1
|
Murphy PV, Dhara A, Fitzgerald LS, Hever E, Konda S, Mandal K. Small lectin ligands as a basis for applications in glycoscience and glycomedicine. Chem Soc Rev 2024; 53:9428-9445. [PMID: 39162695 DOI: 10.1039/d4cs00642a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Glycan recognition by lectins mediates important biological events. This Tutorial Review aims to introduce lectin-ligand interactions and show how these molecular recognition events inspire innovations such as: (i) glycomimetic ligands; (ii) multivalent ligand agonists/antagonists; (iii) ligands for precision delivery of therapies to cells, where therapies include vaccines, siRNA and LYTACs (iv) development of diagnostics. A small number of case studies are selected to demonstrate principles for development of new ligands for applications inspired by knowledge of natural glycan ligand structure and function.
Collapse
Affiliation(s)
- Paul V Murphy
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Ashis Dhara
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
| | - Liam S Fitzgerald
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
- SSPC, SFI Research Centre for Pharmaceuticals, Galway, H91TK33, Ireland
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, University Road, Galway, H91TK33, Ireland
| | - Eoin Hever
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Saidulu Konda
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| | - Kishan Mandal
- School of Biological and Chemical Sciences, Galway, H91TK33, Ireland.
| |
Collapse
|
2
|
Delaunay C, Pollastri S, Thépaut M, Cavazzoli G, Belvisi L, Bouchikri C, Labiod N, Lasala F, Gimeno A, Franconetti A, Jiménez-Barbero J, Ardá A, Delgado R, Bernardi A, Fieschi F. Unprecedented selectivity for homologous lectin targets: differential targeting of the viral receptors L-SIGN and DC-SIGN. Chem Sci 2024:d4sc02980a. [PMID: 39246372 PMCID: PMC11376147 DOI: 10.1039/d4sc02980a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
DC-SIGN (CD209) and L-SIGN (CD209L) are two C-type lectin receptors (CLRs) that facilitate SARS-CoV-2 infections as viral co-receptors. SARS-CoV-2 manipulates both DC-SIGN and L-SIGN for enhanced infection, leading to interest in developing receptor antagonists. Despite their structural similarity (82% sequence identity), they function differently. DC-SIGN, found in dendritic cells, shapes the immune response by recognizing pathogen-associated carbohydrate patterns. In contrast, L-SIGN, expressed in airway epithelial endothelial cells, is not directly involved in immunity. COVID-19's primary threat is the hyperactivation of the immune system, potentially reinforced if DC-SIGN engages with exogenous ligands. Therefore, L-SIGN, co-localized with ACE2-expressing cells in the respiratory tract, is a more suitable target for anti-adhesion therapy. However, designing a selective ligand for L-SIGN is challenging due to the high sequence identity of the Carbohydrate Recognition Domains (CRDs) of the two lectins. We here present Man84, a mannose ring modified with a methylene guanidine triazole at position 2. It binds L-SIGN with a K D of 12.7μM ± 1 μM (ITC) and is the first known L-SIGN selective ligand, showing 50-fold selectivity over DC-SIGN (SPR). The X-ray structure of the L-SIGN CRD/Man84 complex reveals the guanidinium group's role in achieving steric and electrostatic complementarity with L-SIGN. This allows us to trace the source of selectivity to a single amino acid difference between the two CRDs. NMR analysis confirms the binding mode in solution, highlighting Man84's conformational selection upon complex formation. Dimeric versions of Man84 achieve additional selectivity and avidity in the low nanomolar range. These compounds selectively inhibit L-SIGN dependent trans-infection by SARS-CoV-2 and Ebola virus. Man84 and its dimeric constructs display the best affinity and avidity reported to date for low-valency glycomimetics targeting CLRs. They are promising tools for competing with SARS-CoV-2 anchoring in the respiratory tract and have potential for other medical applications.
Collapse
Affiliation(s)
- Clara Delaunay
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale Grenoble France
| | - Sara Pollastri
- Università degli Studi di Milano, Dipartimento di Chimica via Golgi 19 Milano Italy
| | - Michel Thépaut
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale Grenoble France
| | - Gianluca Cavazzoli
- Università degli Studi di Milano, Dipartimento di Chimica via Golgi 19 Milano Italy
| | - Laura Belvisi
- Università degli Studi di Milano, Dipartimento di Chimica via Golgi 19 Milano Italy
| | - Clémentine Bouchikri
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale Grenoble France
| | - Nuria Labiod
- Instituto de Investigacion Hospital Universitario 12 de Octubre, Universidad Complutense, School of Medicine Madrid Spain
| | - Fatima Lasala
- Instituto de Investigacion Hospital Universitario 12 de Octubre, Universidad Complutense, School of Medicine Madrid Spain
| | - Ana Gimeno
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - Antonio Franconetti
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - Jesús Jiménez-Barbero
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science Bilbao Spain
- Centro de Investigacion Biomedica En Red de Enfermedades Respiratorias 28029 Madrid Spain
| | - Ana Ardá
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science Bilbao Spain
| | - Rafael Delgado
- Instituto de Investigacion Hospital Universitario 12 de Octubre, Universidad Complutense, School of Medicine Madrid Spain
- School of Medicine, Universidad Complutense Madrid Spain
| | - Anna Bernardi
- Università degli Studi di Milano, Dipartimento di Chimica via Golgi 19 Milano Italy
| | - Franck Fieschi
- Université Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale Grenoble France
- Institut Universitaire de France (IUF) Paris France
| |
Collapse
|
3
|
Fan CY, Wang SW, Chung C, Chen JY, Chang CY, Chen YC, Hsu TL, Cheng TJR, Wong CH. Synthesis of a dendritic cell-targeted self-assembled polymeric nanoparticle for selective delivery of mRNA vaccines to elicit enhanced immune responses. Chem Sci 2024; 15:11626-11632. [PMID: 39055027 PMCID: PMC11268467 DOI: 10.1039/d3sc06575h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 06/23/2024] [Indexed: 07/27/2024] Open
Abstract
Recent development of SARS-CoV-2 spike mRNA vaccines to control the pandemic is a breakthrough in the field of vaccine development. mRNA vaccines are generally formulated with lipid nanoparticles (LNPs) which are composed of several lipids with specific ratios; however, they generally lack selective delivery. To develop a selective delivery method for mRNA vaccine formulation, we reported here the synthesis of polymeric nanoparticles (PNPs) composed of a guanidine copolymer containing zwitterionic groups and a dendritic cell (DC)-targeted aryl-trimannoside ligand for encapsulation and selective delivery of an mRNA to dendritic cells. A DC-targeted SARS-CoV-2 spike mRNA-PNP vaccine was shown to elicit a stronger protective immune response in mice compared to the traditional mRNA-LNP vaccine and those without the selective delivery design. It is anticipated that this technology is generally applicable to other mRNA vaccines for DC-targeted delivery with enhanced immune response.
Collapse
Affiliation(s)
- Chen-Yo Fan
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Szu-Wen Wang
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Cinya Chung
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Jia-Yan Chen
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Chia-Yen Chang
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Yu-Chen Chen
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | - Tsui-Ling Hsu
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
| | | | - Chi-Huey Wong
- Genomics Research Center, Academia Sinica Taipei 115 Taiwan
- Department of Chemistry, The Scripps Research Institute La Jolla California 92037 USA
| |
Collapse
|
4
|
Lefèbre J, Falk T, Ning Y, Rademacher C. Secondary Sites of the C-type Lectin-Like Fold. Chemistry 2024; 30:e202400660. [PMID: 38527187 DOI: 10.1002/chem.202400660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 03/27/2024]
Abstract
C-type lectins are a large superfamily of proteins involved in a multitude of biological processes. In particular, their involvement in immunity and homeostasis has rendered them attractive targets for diverse therapeutic interventions. They share a characteristic C-type lectin-like domain whose adaptability enables them to bind a broad spectrum of ligands beyond the originally defined canonical Ca2+-dependent carbohydrate binding. Together with variable domain architecture and high-level conformational plasticity, this enables C-type lectins to meet diverse functional demands. Secondary sites provide another layer of regulation and are often intricately linked to functional diversity. Located remote from the canonical primary binding site, secondary sites can accommodate ligands with other physicochemical properties and alter protein dynamics, thus enhancing selectivity and enabling fine-tuning of the biological response. In this review, we outline the structural determinants allowing C-type lectins to perform a large variety of tasks and to accommodate the ligands associated with it. Using the six well-characterized Ca2+-dependent and Ca2+-independent C-type lectin receptors DC-SIGN, langerin, MGL, dectin-1, CLEC-2 and NKG2D as examples, we focus on the characteristics of non-canonical interactions and secondary sites and their potential use in drug discovery endeavors.
Collapse
Affiliation(s)
- Jonathan Lefèbre
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Torben Falk
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Yunzhan Ning
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Vienna Doctoral School of Pharmaceutical, Nutritional and Sport, Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
- Department of Microbiology, Immunology and Genetics, University of Vienna, Max F. Perutz Labs, Vienna, Austria
| |
Collapse
|
5
|
Basaran R, Ning X, Budhadev D, Hondow N, Guo Y, Zhou D. Probing the pH-dependency of DC-SIGN/R multivalent lectin-glycan interactions using polyvalent glycan-gold nanoparticles. NANOSCALE ADVANCES 2024; 6:2198-2208. [PMID: 38633047 PMCID: PMC11019501 DOI: 10.1039/d3na01013a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024]
Abstract
The dendritic cell tetrameric lectin, DC-SIGN, and its closely related endothelial cell lectin, DC-SIGNR (collectively abbreviated as DC-SIGN/R) play a key role in the binding and transmission of deadly viruses, including Ebola, HIV, HCV, and SARS-CoV-2. Their virus binding/release processes involve a gradually acidifying environment following the natural intracellular trafficking pathways. Therefore, understanding DC-SIGN/R's pH-dependent binding properties with glycan ligands is of great importance. We have recently developed densely glycosylated gold nanoparticles (glycan-GNPs) as a powerful new tool for probing DC-SIGN/R multivalent lectin-glycan interaction (MLGI) mechanisms. They can provide not only quantitative MLGI affinities but also important structural information, such as binding site orientation and binding modes. Herein, we further employ the glycan-GNP probes to investigate the pH dependency of DC-SIGN/R MLGI properties. We find that DC-SIGN/R MLGIs exhibit distinct pH dependence over the normal physiological (7.4) to lysosomal (∼4.6) pH range. DC-SIGN binds glycan-GNPs strongly and stably from pH 7.4 to ∼5.8, but the binding is weakened significantly as pH decreases to ≤5.4 and may be fully dissociated at pH 4.6. This behaviour is fully consistent with DC-SIGN's role as an endocytic recycling receptor. In contrast, DC-SIGNR's affinity with glycan-GNPs is enhanced with the decreasing pH from 7.4 to 5.4, peaking at pH 5.4, and then reduced as pH is further lowered. Interestingly, both DC-SIGN/R binding with glycan-GNPs are found to be partially reversible in a pH-dependent manner.
Collapse
Affiliation(s)
- Rahman Basaran
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Xinyu Ning
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Darshita Budhadev
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds Leeds LS2 9JT UK
| | - Yuan Guo
- School of Food Science and Nutrition, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| | - Dejian Zhou
- School of Chemistry, Astbury Centre for Structural Molecular Biology, University of Leeds Leeds LS2 9JT UK
| |
Collapse
|
6
|
Nieto-Fabregat F, Zhu Q, Vivès C, Zhang Y, Marseglia A, Chiodo F, Thépaut M, Rai D, Kulkarni SS, Di Lorenzo F, Molinaro A, Marchetti R, Fieschi F, Xiao G, Yu B, Silipo A. Atomic-Level Dissection of DC-SIGN Recognition of Bacteroides vulgatus LPS Epitopes. JACS AU 2024; 4:697-712. [PMID: 38425910 PMCID: PMC10900495 DOI: 10.1021/jacsau.3c00748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 03/02/2024]
Abstract
The evaluation of Bacteroides vulgatus mpk (BVMPK) lipopolysaccharide (LPS) recognition by DC-SIGN, a key lectin in mediating immune homeostasis, has been here performed. A fine chemical dissection of BVMPK LPS components, attained by synthetic chemistry combined to spectroscopic, biophysical, and computational techniques, allowed to finely map the LPS epitopes recognized by DC-SIGN. Our findings reveal BVMPK's role in immune modulation via DC-SIGN, targeting both the LPS O-antigen and the core oligosaccharide. Furthermore, when framed within medical chemistry or drug design, our results could lead to the development of tailored molecules to benefit the hosts dealing with inflammatory diseases.
Collapse
Affiliation(s)
- Ferran Nieto-Fabregat
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Qian Zhu
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai
Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Corinne Vivès
- Université
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble 38027, France
| | - Yunqin Zhang
- State Key
Laboratory of Phytochemistry and Plant Resources in West China, Kunming
Institute of Botany, University of Chinese
Academy of Sciences, Chinese Academy of Sciences, Kunming 650201, China
| | - Angela Marseglia
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Fabrizio Chiodo
- Institute
of Biomolecular Chemistry, National Research Council (CNR), Pozzuoli 80078, Italy
| | - Michel Thépaut
- Université
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble 38027, France
| | - Diksha Rai
- Department
of Chemistry, Indian Institute of Technology
Bombay, Powai, Mumbai 400076, India
| | - Suvarn S. Kulkarni
- Department
of Chemistry, Indian Institute of Technology
Bombay, Powai, Mumbai 400076, India
| | - Flaviana Di Lorenzo
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Antonio Molinaro
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Roberta Marchetti
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| | - Franck Fieschi
- Université
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble 38027, France
- Institut
Universitaire de France (IUF), Paris 75005, France
| | - Guozhi Xiao
- State Key
Laboratory of Phytochemistry and Plant Resources in West China, Kunming
Institute of Botany, University of Chinese
Academy of Sciences, Chinese Academy of Sciences, Kunming 650201, China
| | - Biao Yu
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai
Institute of Organic Chemistry, University
of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Alba Silipo
- Department
of Chemical Sciences, University of Naples
Federico II, Naples 80126, Italy
| |
Collapse
|
7
|
Niveau C, Sosa Cuevas E, Roubinet B, Pezet M, Thépaut M, Mouret S, Charles J, Fieschi F, Landemarre L, Chaperot L, Saas P, Aspord C. Melanoma tumour-derived glycans hijack dendritic cell subsets through C-type lectin receptor binding. Immunology 2024; 171:286-311. [PMID: 37991344 DOI: 10.1111/imm.13717] [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: 07/07/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023] Open
Abstract
Dendritic cell (DC) subsets play a crucial role in shaping anti-tumour immunity. Cancer escapes from the control immune system by hijacking DC functions. Yet, bases for such subversion are only partially understood. Tumour cells display aberrant glycan motifs on surface glycoproteins and glycolipids. Such carbohydrate patterns can be sensed by DCs through C-type lectin receptors (CLRs) that are critical to shape and orientate immune responses. We recently demonstrated that melanoma tumour cells harboured an aberrant 'glyco-code,' and that circulating and tumour-infiltrating DCs from melanoma patients displayed major perturbations in their CLR profiles. To decipher whether melanoma, through aberrant glycan patterns, may exploit CLR pathways to mislead DCs and evade immune control, we explored the impact of glycan motifs aberrantly found in melanoma (neoglycoproteins [NeoGP] functionalised with Gal, Man, GalNAc, s-Tn, fucose [Fuc] and GlcNAc residues) on features of human DC subsets (cDC2s, cDC1s and pDCs). We examined the ability of glycans to bind to purified DCs, and assessed their impact on DC basal properties and functional features using flow cytometry, confocal microscopy and multiplex secreted protein analysis. DC subsets differentially bound and internalised NeoGP depending on the nature of the glycan. Strikingly, Fuc directly remodelled the expression of activation markers and immune checkpoints, as well as the cytokine/chemokine secretion profile of DC subsets. NeoGP interfered with Toll like receptor (TLR)-signalling and pre-conditioned DCs to exhibit an altered response to subsequent TLR stimulation, dampening antitumor mediators while triggering pro-tumoral factors. We further demonstrated that DC subsets can bind NeoGP through CLRs, and identified GalNAc/MGL and s-Tn/ C-type lectin-like receptor 2 (CLEC2) as potential candidates. Moreover, DC dysfunction induced by tumour-associated carbohydrate molecules may be reversed by interfering with the glycan/CLR axis. These findings revealed the glycan/CLR axis as a promising checkpoint to exploit in order to reshape potent antitumor immunity while impeding immunosuppressive pathways triggered by aberrant tumour glycosylation patterns. This may rescue DCs from tumour hijacking and improve clinical success in cancer patients.
Collapse
Affiliation(s)
- Camille Niveau
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Eleonora Sosa Cuevas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | | | - Mylène Pezet
- Institute for Advanced Biosciences, Plateforme de Microscopie Photonique-Imagerie Cellulaire et Cytométrie en Flux (Microcell), Inserm U1209-CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Michel Thépaut
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Stéphane Mouret
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Julie Charles
- Dermatology, Allergology & Photobiology Department, CHU Grenoble Alpes, Université Grenoble Alpes, Grenoble, France
| | - Franck Fieschi
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | | | - Laurence Chaperot
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Philippe Saas
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| | - Caroline Aspord
- Institute for Advanced Biosciences, Team: Epigenetics, Immunity, Metabolism, Cell Signaling & Cancer, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
- R&D Laboratory, Etablissement Français du Sang Auvergne-Rhône-Alpes, Grenoble, France
| |
Collapse
|
8
|
Herrera-González I, González-Cuesta M, Thépaut M, Laigre E, Goyard D, Rojo J, García Fernández JM, Fieschi F, Renaudet O, Nieto PM, Ortiz Mellet C. High-Mannose Oligosaccharide Hemimimetics that Recapitulate the Conformation and Binding Mode to Concanavalin A, DC-SIGN and Langerin. Chemistry 2024; 30:e202303041. [PMID: 37828571 DOI: 10.1002/chem.202303041] [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: 09/18/2023] [Revised: 10/12/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
The "carbohydrate chemical mimicry" exhibited by sp2 -iminosugars has been utilized to develop practical syntheses for analogs of the branched high-mannose-type oligosaccharides (HMOs) Man3 and Man5 . In these compounds, the terminal nonreducing Man residues have been substituted with 5,6-oxomethylidenemannonojirimycin (OMJ) motifs. The resulting oligomannoside hemimimetic accurately reproduce the structure, configuration, and conformational behavior of the original mannooligosaccharides, as confirmed by NMR and computational techniques. Binding studies with mannose binding lectins, including concanavalin A, DC-SIGN, and langerin, by enzyme-linked lectin assay and surface plasmon resonance revealed significant variations in their ability to accommodate the OMJ unit in the mannose binding site. Intriguingly, OMJMan segments demonstrated "in line" heteromultivalent effects during binding to the three lectins. Similar to the mannobiose (Man2 ) branches in HMOs, the binding modes involving the external or internal monosaccharide unit at the carbohydrate binding-domain exist in equilibrium, facilitating sliding and recapture processes. This equilibrium, which influences the multivalent binding of HMOs, can be finely modulated upon incorporation of the OMJ sp2 -iminosugar caps. As a proof of concept, the affinity and selectivity towards DC-SIGN and langerin were adjustable by presenting the OMJMan epitope in platforms with diverse architectures and valencies.
Collapse
Affiliation(s)
- Irene Herrera-González
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
- Present address: DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Manuel González-Cuesta
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
| | - Michel Thépaut
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
| | - Eugénie Laigre
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - David Goyard
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Javier Rojo
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - José M García Fernández
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - Franck Fieschi
- Institut de Biologie Structurale, Université Grenoble Alpes, CNRS, CEA, 38000, Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | - Olivier Renaudet
- DCM, UMR 5250, Université Grenoble Alpes, CNRS, 570 Rue de la Chimie, 38000, Grenoble, France
| | - Pedro M Nieto
- Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Américo Vespucio 49, 41092, Sevilla, Spain
| | - Carmen Ortiz Mellet
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, C/ Profesor García González 1, 41012, Sevilla, Spain
| |
Collapse
|
9
|
Bains A, Fischer K, Guan W, LiWang PJ. The Antiviral Activity of the Lectin Griffithsin against SARS-CoV-2 Is Enhanced by the Presence of Structural Proteins. Viruses 2023; 15:2452. [PMID: 38140693 PMCID: PMC10747160 DOI: 10.3390/v15122452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/07/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Although COVID-19 transmission has been reduced by the advent of vaccinations and a variety of rapid monitoring techniques, the SARS-CoV-2 virus itself has shown a remarkable ability to mutate and persist. With this long track record of immune escape, researchers are still exploring prophylactic treatments to curtail future SARS-CoV-2 variants. Specifically, much focus has been placed on the antiviral lectin Griffithsin in preventing spike protein-mediated infection via the hACE2 receptor (direct infection). However, an oft-overlooked aspect of SARS-CoV-2 infection is viral capture by attachment receptors such as DC-SIGN, which is thought to facilitate the initial stages of COVID-19 infection in the lung tissue (called trans-infection). In addition, while immune escape is dictated by mutations in the spike protein, coronaviral virions also incorporate M, N, and E structural proteins within the particle. In this paper, we explored how several structural facets of both the SARS-CoV-2 virion and the antiviral lectin Griffithsin can affect and attenuate the infectivity of SARS-CoV-2 pseudovirus. We found that Griffithsin was a better inhibitor of hACE2-mediated direct infection when the coronaviral M protein is present compared to when it is absent (possibly providing an explanation regarding why Griffithsin shows better inhibition against authentic SARS-CoV-2 as opposed to pseudotyped viruses, which generally do not contain M) and that Griffithsin was not an effective inhibitor of DC-SIGN-mediated trans-infection. Furthermore, we found that DC-SIGN appeared to mediate trans-infection exclusively via binding to the SARS-CoV-2 spike protein, with no significant effect observed when other viral proteins (M, N, and/or E) were present. These results provide etiological data that may help to direct the development of novel antiviral treatments, either by leveraging Griffithsin binding to the M protein as a novel strategy to prevent SARS-CoV-2 infection or by narrowing efforts to inhibit trans-infection to focus on DC-SIGN binding to SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- Arjan Bains
- Chemistry and Biochemistry, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Kathryn Fischer
- Quantitative and Systems Biology, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Wenyan Guan
- Materials and Biomaterials Science and Engineering, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Patricia J. LiWang
- Molecular Cell Biology, Health Sciences Research Institute, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| |
Collapse
|
10
|
Bains A, Guan W, LiWang PJ. The Effect of Select SARS-CoV-2 N-Linked Glycan and Variant of Concern Spike Protein Mutations on C-Type Lectin-Receptor-Mediated Infection. Viruses 2023; 15:1901. [PMID: 37766307 PMCID: PMC10535197 DOI: 10.3390/v15091901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
The SARS-CoV-2 virion has shown remarkable resilience, capable of mutating to escape immune detection and re-establishing infectious capabilities despite new vaccine rollouts. Therefore, there is a critical need to identify relatively immutable epitopes on the SARS-CoV-2 virion that are resistant to future mutations the virus may accumulate. While hACE2 has been identified as the receptor that mediates SARS-CoV-2 susceptibility, it is only modestly expressed in lung tissue. C-type lectin receptors like DC-SIGN can act as attachment sites to enhance SARS-CoV-2 infection of cells with moderate or low hACE2 expression. We developed an easy-to-implement assay system that allows for the testing of SARS-CoV-2 trans-infection. Using our assay, we assessed how SARS-CoV-2 Spike S1-domain glycans and spike proteins from different strains affected the ability of pseudotyped lentivirions to undergo DC-SIGN-mediated trans-infection. Through our experiments with seven glycan point mutants, two glycan cluster mutants and four strains of SARS-CoV-2 spike, we found that glycans N17 and N122 appear to have significant roles in maintaining COVID-19's infectious capabilities. We further found that the virus cannot retain infectivity upon the loss of multiple glycosylation sites, and that Omicron BA.2 pseudovirions may have an increased ability to bind to other non-lectin receptor proteins on the surface of cells. Taken together, our work opens the door to the development of new therapeutics that can target overlooked epitopes of the SARS-CoV-2 virion to prevent C-type lectin-receptor-mediated trans-infection in lung tissue.
Collapse
Affiliation(s)
- Arjan Bains
- Chemistry and Biochemistry, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Wenyan Guan
- Materials and Biomaterials Science and Engineering, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA;
| | - Patricia J. LiWang
- Molecular Cell Biology, Health Sciences Research Institute, University of California Merced, 5200 North Lake Rd., Merced, CA 95343, USA
| |
Collapse
|
11
|
Viljoen A, Vercellone A, Chimen M, Gaibelet G, Mazères S, Nigou J, Dufrêne YF. Nanoscale clustering of mycobacterial ligands and DC-SIGN host receptors are key determinants for pathogen recognition. SCIENCE ADVANCES 2023; 9:eadf9498. [PMID: 37205764 PMCID: PMC10198640 DOI: 10.1126/sciadv.adf9498] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
The bacterial pathogen Mycobacterium tuberculosis binds to the C-type lectin DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) on dendritic cells to evade the immune system. While DC-SIGN glycoconjugate ligands are ubiquitous among mycobacterial species, the receptor selectively binds pathogenic species from the M. tuberculosis complex (MTBC). Here, we unravel the molecular mechanism behind this intriguing selective recognition by means of a multidisciplinary approach combining single-molecule atomic force microscopy with Förster resonance energy transfer and bioassays. Molecular recognition imaging of mycobacteria demonstrates that the distribution of DC-SIGN ligands markedly differs between Mycobacterium bovis Bacille Calmette-Guérin (BCG) (model MTBC species) and Mycobacterium smegmatis (non-MTBC species), the ligands being concentrated into dense nanodomains on M. bovis BCG. Upon bacteria-host cell adhesion, ligand nanodomains induce the recruitment and clustering of DC-SIGN. Our study highlights the key role of clustering of both ligands on MTBC species and DC-SIGN host receptors in pathogen recognition, a mechanism that might be widespread in host-pathogen interactions.
Collapse
Affiliation(s)
- Albertus Viljoen
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07., B-1348 Louvain-la-Neuve, Belgium
| | - Alain Vercellone
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Myriam Chimen
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Gérald Gaibelet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Jérôme Nigou
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Yves F. Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Croix du Sud, 4-5, bte L7.07.07., B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
12
|
Porkolab V, Lepšík M, Ordanini S, St John A, Le Roy A, Thépaut M, Paci E, Ebel C, Bernardi A, Fieschi F. Powerful Avidity with a Limited Valency for Virus-Attachment Blockers on DC-SIGN: Combining Chelation and Statistical Rebinding with Structural Plasticity of the Receptor. ACS CENTRAL SCIENCE 2023; 9:709-718. [PMID: 37122470 PMCID: PMC10141607 DOI: 10.1021/acscentsci.2c01136] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Indexed: 05/03/2023]
Abstract
The C-type lectin receptor DC-SIGN has been highlighted as the coreceptor for the spike protein of the SARS-CoV-2 virus. A multivalent glycomimetic ligand, Polyman26, has been found to inhibit DC-SIGN-dependent trans-infection of SARS-CoV-2. The molecular details underlying avidity generation in such systems remain poorly characterized. In an effort to dissect the contribution of the known multivalent effects - chelation, clustering, and statistical rebinding - we studied a series of dendrimer constructs related to Polyman26 with a rod core rationally designed to engage simultaneously two binding sites of the tetrameric DC-SIGN. Binding properties of these compounds have been studied with a range of biophysical techniques, including recently developed surface plasmon resonance oriented-surface methodology. Using molecular modeling we addressed, for the first time, the impact of the carbohydrate recognition domains' flexibility of the DC-SIGN tetramer on the compounds' avidity. We were able to gain deeper insight into the role of different binding modes, which in combination produce a construct with a nanomolar affinity despite a limited valency. This multifaceted experimental-theoretical approach provides detailed understanding of multivalent ligand/multimeric protein interactions which can lead to future predictions. This work opens the way to the development of new virus attachment blockers adapted to different C-type lectin receptors of viruses.
Collapse
Affiliation(s)
- Vanessa Porkolab
- Univ.
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Martin Lepšík
- Univ.
Grenoble Alpes, CNRS, CERMAV, 38000 Grenoble, France
- Institute
of Organic Chemistry and Biochemistry, Czech
Academy of Sciences, Flemingovo nam. 2, Prague 6, 166 10, Czechia
| | - Stefania Ordanini
- Universita’
degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, 20133, Milano, Italy
| | - Alexander St John
- Astbury
Centre & School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Aline Le Roy
- Univ.
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Michel Thépaut
- Univ.
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Emanuele Paci
- Department
of Physics and Astronomy “Augusto Righi”, University of Bologna, Via Zamboni, 33, 40126, Bologna, Italy
| | - Christine Ebel
- Univ.
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Anna Bernardi
- Universita’
degli Studi di Milano, Dipartimento di Chimica, via Golgi 19, 20133, Milano, Italy
| | - Franck Fieschi
- Univ.
Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
- Institut
Universitaire de France (IUF), 1 rue Descartes, 75231 Paris, France
| |
Collapse
|
13
|
Hooper J, Budhadev D, Fernandez Ainaga DL, Hondow N, Zhou D, Guo Y. Polyvalent Glycan Functionalized Quantum Nanorods as Mechanistic Probes for Shape-Selective Multivalent Lectin-Glycan Recognition. ACS APPLIED NANO MATERIALS 2023; 6:4201-4213. [PMID: 37006911 PMCID: PMC10043877 DOI: 10.1021/acsanm.2c05247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Multivalent lectin-glycan interactions (MLGIs) are widespread in biology and hold the key to many therapeutic applications. However, the underlying structural and biophysical mechanisms for many MLGIs remain poorly understood, limiting our ability to design glycoconjugates to potently target specific MLGIs for therapeutic intervention. Glycosylated nanoparticles have emerged as a powerful biophysical probe for MLGIs, although how nanoparticle shape affects the MLGI molecular mechanisms remains largely unexplored. Herein, we have prepared fluorescent quantum nanorods (QRs), densely coated with α-1,2-manno-biose ligands (QR-DiMan), as multifunctional probes to investigate how scaffold geometry affects the MLGIs of a pair of closely related, tetrameric viral receptors, DC-SIGN and DC-SIGNR. We have previously shown that a DiMan-capped spherical quantum dot (QD-DiMan) gives weak cross-linking interactions with DC-SIGNR but strong simultaneous binding with DC-SIGN. Against the elongated QR-DiMan, DC-SIGN retains similarly strong simultaneous binding of all four binding sites with a single QR-DiMan (apparent K d ≈ 0.5 nM, ∼1.8 million-fold stronger than the corresponding monovalent binding), while DC-SIGNR gives both weak cross-linking and strong individual binding interactions, resulting in a larger binding affinity enhancement than that with QD-DiMan. S/TEM analysis of QR-DiMan-lectin assemblies reveals that DC-SIGNR's different binding modes arise from the different nanosurface curvatures of the QR scaffold. The glycan display at the spherical ends presents too high a steric barrier for DC-SIGNR to bind with all four binding sites; thus, it cross-links between two QR-DiMan to maximize binding multivalency, whereas the more planar character of the cylindrical center allows the glycans to bridge all binding sites in DC-SIGNR. This work thus establishes glycosylated QRs as a powerful biophysical probe for MLGIs not only to provide quantitative binding affinities and binding modes but also to demonstrate the specificity of multivalent lectins in discriminating different glycan displays in solution, dictated by the scaffold curvature.
Collapse
Affiliation(s)
- James Hooper
- School
of Food Science and Nutrition and Astbury Centre for Structural Molecular
Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Darshita Budhadev
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | - Nicole Hondow
- School
of Chemical and Process Engineering, University
of Leeds, Leeds LS2 9JT, United
Kingdom
| | - Dejian Zhou
- School
of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Yuan Guo
- School
of Food Science and Nutrition and Astbury Centre for Structural Molecular
Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
14
|
Martínez-Bailén M, Rojo J, Ramos-Soriano J. Multivalent glycosystems for human lectins. Chem Soc Rev 2023; 52:536-572. [PMID: 36545903 DOI: 10.1039/d2cs00736c] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human lectins are involved in a wide variety of biological processes, both physiological and pathological, which have attracted the interest of the scientific community working in the glycoscience field. Multivalent glycosystems have been employed as useful tools to understand carbohydrate-lectin binding processes as well as for biomedical applications. The review shows the different scaffolds designed for a multivalent presentation of sugars and their corresponding binding studies to lectins and in some cases, their biological activities. We summarise this research by organizing based on lectin types to highlight the progression in this active field. The paper provides an overall picture of how these contributions have furnished relevant information on this topic to help in understanding and participate in these carbohydrate-lectin interactions.
Collapse
Affiliation(s)
- Macarena Martínez-Bailén
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| | - Javier Ramos-Soriano
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, Seville 41092, Spain.
| |
Collapse
|
15
|
McLeish KR, Fernandes MJ. Understanding inhibitory receptor function in neutrophils through the lens of
CLEC12A. Immunol Rev 2022; 314:50-68. [PMID: 36424898 DOI: 10.1111/imr.13174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Neutrophils are the first leukocytes recruited from the circulation in response to invading pathogens or injured cells. To eradicate pathogens and contribute to tissue repair, recruited neutrophils generate and release a host of toxic chemicals that can also damage normal cells. To avoid collateral damage leading to tissue injury and organ dysfunction, molecular mechanisms evolved that tightly control neutrophil response threshold to activating signals, the strength and location of the response, and the timing of response termination. One mechanism of response control is interruption of activating intracellular signaling pathways by the 20 inhibitory receptors expressed by neutrophils. The two inhibitory C-type lectin receptors expressed by neutrophils, CLEC12A and DCIR, exhibit both common and distinct molecular and functional mechanisms, and they are associated with different diseases. In this review, we use studies on CLEC12A as a model of inhibitory receptor regulation of neutrophil function and participation in disease. Understanding the molecular mechanisms leading to inhibitory receptor specificity offers the possibility of using physiologic control of neutrophil functions as a pharmacologic tool to control inflammatory diseases.
Collapse
Affiliation(s)
- Kenneth R. McLeish
- Department of Medicine University of Louisville School of Medicine Louisville Kentucky USA
| | - Maria J. Fernandes
- Infectious and Immune Diseases Division CHU de Québec‐Laval University Research Center Québec Québec Canada
- Department of Microbiology‐Infectious Diseases and Immunology, Faculty of Medicine Laval University Québec Québec Canada
| |
Collapse
|
16
|
Ramos-Soriano J, Illescas BM, Pérez-Sánchez A, Sánchez-Bento R, Lasala F, Rojo J, Delgado R, Martín N. Topological and Multivalent Effects in Glycofullerene Oligomers as EBOLA Virus Inhibitors. Int J Mol Sci 2022; 23:ijms23095083. [PMID: 35563489 PMCID: PMC9131134 DOI: 10.3390/ijms23095083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/25/2022] Open
Abstract
The synthesis of new biocompatible antiviral materials to fight against the development of multidrug resistance is being widely explored. Due to their unique globular structure and excellent properties, [60]fullerene-based antivirals are very promising bioconjugates. In this work, fullerene derivatives with different topologies and number of glycofullerene units were synthesized by using a SPAAC copper free strategy. This procedure allowed the synthesis of compounds 1–3, containing from 20 to 40 mannose units, in a very efficient manner and in short reaction times under MW irradiation. The glycoderivatives were studied in an infection assay by a pseudotyped viral particle with Ebola virus GP1. The results obtained show that these glycofullerene oligomers are efficient inhibitors of EBOV infection with IC50s in the nanomolar range. In particular, compound 3, with four glycofullerene moieties, presents an outstanding relative inhibitory potency (RIP). We propose that this high RIP value stems from the appropriate topological features that efficiently interact with DC-SIGN.
Collapse
Affiliation(s)
- Javier Ramos-Soriano
- Departamento de Química Orgánica, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain; (J.R.-S.); (A.P.-S.); (R.S.-B.); (N.M.)
| | - Beatriz M. Illescas
- Departamento de Química Orgánica, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain; (J.R.-S.); (A.P.-S.); (R.S.-B.); (N.M.)
- Correspondence: (B.M.I.); (R.D.)
| | - Alfonso Pérez-Sánchez
- Departamento de Química Orgánica, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain; (J.R.-S.); (A.P.-S.); (R.S.-B.); (N.M.)
| | - Raquel Sánchez-Bento
- Departamento de Química Orgánica, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain; (J.R.-S.); (A.P.-S.); (R.S.-B.); (N.M.)
| | - Fátima Lasala
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC–Universidad de Sevilla, Av. Américo Vespucio 49, 41092 Seville, Spain;
| | - Rafael Delgado
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain;
- Correspondence: (B.M.I.); (R.D.)
| | - Nazario Martín
- Departamento de Química Orgánica, Facultad de Química, Universidad Complutense, 28040 Madrid, Spain; (J.R.-S.); (A.P.-S.); (R.S.-B.); (N.M.)
- IMDEA-Nanoscience, C/Faraday, 9, Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
17
|
Goti G, Colombo C, Achilli S, Vives C, Thépaut M, Luczkowiak J, Labiod N, Delgado R, Fieschi F, Bernardi A, Vivès C. Precision Glycodendrimers for DC‐SIGN Targeting. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Giulio Goti
- Università degli Studi di Milano: Universita degli Studi di Milano Chemistry ITALY
| | - Cinzia Colombo
- Università degli Studi di Milano: Universita degli Studi di Milano Chemistry ITALY
| | | | | | | | - Joanna Luczkowiak
- Hospital Universitario 12 de Octubre Instituto de Investigación SPAIN
| | - Nuria Labiod
- Hospital Universitario 12 de Octubre Instituto de Investigación SPAIN
| | - Rafael Delgado
- Hospital Universitario 12 de Octubre Instituto de Investigación SPAIN
| | | | - Anna Bernardi
- Universita' di Milano Chimica via Golgi 19 20133 Milano ITALY
| | | |
Collapse
|
18
|
Fischer S, Stegmann F, Gnanapragassam VS, Lepenies B. From structure to function – Ligand recognition by myeloid C-type lectin receptors. Comput Struct Biotechnol J 2022; 20:5790-5812. [DOI: 10.1016/j.csbj.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022] Open
|
19
|
Ambati S, Pham T, Lewis ZA, Lin X, Meagher RB. DC-SIGN targets amphotericin B-loaded liposomes to diverse pathogenic fungi. Fungal Biol Biotechnol 2021; 8:22. [PMID: 34952645 PMCID: PMC8709943 DOI: 10.1186/s40694-021-00126-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Life-threatening invasive fungal infections are treated with antifungal drugs such as Amphotericin B (AmB) loaded liposomes. Our goal herein was to show that targeting liposomal AmB to fungal cells with the C-type lectin pathogen recognition receptor DC-SIGN improves antifungal activity. DC-SIGN binds variously crosslinked mannose-rich and fucosylated glycans and lipomannans that are expressed by helminth, protist, fungal, bacterial and viral pathogens including three of the most life-threatening fungi, Aspergillus fumigatus, Candida albicans and Cryptococcus neoformans. Ligand recognition by human DC-SIGN is provided by a carbohydrate recognition domain (CRD) linked to the membrane transit and signaling sequences. Different combinations of the eight neck repeats (NR1 to NR8) expressed in different protein isoforms may alter the orientation of the CRD to enhance its binding to different glycans. RESULTS We prepared two recombinant isoforms combining the CRD with NR1 and NR2 in isoform DCS12 and with NR7 and NR8 in isoform DCS78 and coupled them to a lipid carrier. These constructs were inserted into the membrane of pegylated AmB loaded liposomes AmB-LLs to produce DCS12-AmB-LLs and DCS78-AmB-LLs. Relative to AmB-LLs and Bovine Serum Albumin coated BSA-AmB-LLs, DCS12-AmB-LLs and DCS78-AmB-LLs bound more efficiently to the exopolysaccharide matrices produced by A. fumigatus, C. albicans and C. neoformans in vitro, with DCS12-AmB-LLs performing better than DCS78-AmB-LLs. DCS12-AmB-LLs inhibited and/or killed all three species in vitro significantly better than AmB-LLs or BSA-AmB-LLs. In mouse models of invasive candidiasis and pulmonary aspergillosis, one low dose of DCS12-AmB-LLs significantly reduced the fungal burden in the kidneys and lungs, respectively, several-fold relative to AmB-LLs. CONCLUSIONS DC-SIGN's CRD specifically targeted antifungal liposomes to three highly evolutionarily diverse pathogenic fungi and enhanced the antifungal efficacy of liposomal AmB both in vitro and in vivo. Targeting significantly reduced the effective dose of antifungal drug, which may reduce drug toxicity, be effective in overcoming dose dependent drug resistance, and more effectively kill persister cells. In addition to fungi, DC-SIGN targeting of liposomal packaged anti-infectives have the potential to alter treatment paradigms for a wide variety of pathogens from different kingdoms including protozoans, helminths, bacteria, and viruses which express its cognate ligands.
Collapse
Affiliation(s)
- Suresh Ambati
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA.
| | - Tuyetnhu Pham
- Department of Microbiology, University of Georgia, Athens, GA, 30602, USA
| | - Zachary A Lewis
- Department of Microbiology, University of Georgia, Athens, GA, 30602, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, GA, 30602, USA
| | - Richard B Meagher
- Department of Genetics, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
20
|
Cramer J. Medicinal chemistry of the myeloid C-type lectin receptors Mincle, Langerin, and DC-SIGN. RSC Med Chem 2021; 12:1985-2000. [PMID: 35024612 PMCID: PMC8672822 DOI: 10.1039/d1md00238d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/14/2021] [Indexed: 01/07/2023] Open
Abstract
In their role as pattern-recognition receptors on cells of the innate immune system, myeloid C-type lectin receptors (CLRs) assume important biological functions related to immunity, homeostasis, and cancer. As such, this family of receptors represents an appealing target for therapeutic interventions for modulating the outcome of many pathological processes, in particular related to infectious diseases. This review summarizes the current state of research into glycomimetic or drug-like small molecule ligands for the CLRs Mincle, Langerin, and DC-SIGN, which have potential therapeutic applications in vaccine research and anti-infective therapy.
Collapse
Affiliation(s)
- Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University of Düsseldorf Universitätsstr. 1 40225 Düsseldorf Germany
| |
Collapse
|
21
|
Carbohydrates from Pseudomonas aeruginosa biofilms interact with immune C-type lectins and interfere with their receptor function. NPJ Biofilms Microbiomes 2021; 7:87. [PMID: 34880222 PMCID: PMC8655052 DOI: 10.1038/s41522-021-00257-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 11/03/2021] [Indexed: 11/16/2022] Open
Abstract
Bacterial biofilms represent a challenge to the healthcare system because of their resilience against antimicrobials and immune attack. Biofilms consist of bacterial aggregates embedded in an extracellular polymeric substance (EPS) composed of polysaccharides, nucleic acids and proteins. We hypothesised that carbohydrates could contribute to immune recognition of Pseudomonas aeruginosa biofilms by engaging C-type lectins. Here we show binding of Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin (DC-SIGN, CD209), mannose receptor (MR, CD206) and Dectin-2 to P. aeruginosa biofilms. We also demonstrate that DC-SIGN, unlike MR and Dectin-2, recognises planktonic P. aeruginosa cultures and this interaction depends on the presence of the common polysaccharide antigen. Within biofilms DC-SIGN, Dectin-2 and MR ligands appear as discrete clusters with dispersed DC-SIGN ligands also found among bacterial aggregates. DC-SIGN, MR and Dectin-2 bind to carbohydrates purified from P. aeruginosa biofilms, particularly the high molecular weight fraction (HMW; >132,000 Da), with KDs in the nM range. These HMW carbohydrates contain 74.9–80.9% mannose, display α-mannan segments, interfere with the endocytic activity of cell-associated DC-SIGN and MR and inhibit Dectin-2-mediated cellular activation. In addition, biofilm carbohydrates reduce the association of the DC-SIGN ligand Lewisx, but not fucose, to human monocyte-derived dendritic cells (moDCs), and alter moDC morphology without affecting early cytokine production in response to lipopolysaccharide or P. aeruginosa cultures. This work identifies the presence of ligands for three important C-type lectins within P. aeruginosa biofilm structures and purified biofilm carbohydrates and highlights the potential for these receptors to impact immunity to P. aeruginosa infection.
Collapse
|
22
|
Chakroun K, Taouai M, Porkolab V, Luczkowiak J, Sommer R, Cheneau C, Mathiron D, Ben Maaouia MA, Pilard S, Abidi R, Mullié C, Fieschi F, Cragg PJ, Halary F, Delgado R, Benazza M. Low-Valent Calix[4]arene Glycoconjugates Based on Hydroxamic Acid Bearing Linkers as Potent Inhibitors in a Model of Ebola Virus Cis-Infection and HCMV-gB-Recombinant Glycoprotein Interaction with MDDC Cells by Blocking DC-SIGN. J Med Chem 2021; 64:14332-14343. [PMID: 34524803 DOI: 10.1021/acs.jmedchem.1c00818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In addition to a variety of viral-glycoprotein receptors (e.g., heparan sulfate, Niemann-Pick C1, etc.), dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), from the C-type lectin receptor family, plays one of the most important pathogenic functions for a wide range of viruses (e.g., Ebola, human cytomegalovirus (HCMV), HIV-1, severe acute respiratory syndrome coronavirus 2, etc.) that invade host cells before replication; thus, its inhibition represents a relevant extracellular antiviral therapy. We report two novel p-tBu-calixarene glycoclusters 1 and 2, bearing tetrahydroxamic acid groups, which exhibit micromolar inhibition of soluble DC-SIGN binding and provide nanomolar IC50 inhibition of both DC-SIGN-dependent Jurkat cis-cell infection by viral particle pseudotyped with Ebola virus glycoprotein and the HCMV-gB-recombinant glycoprotein interaction with monocyte-derived dendritic cells expressing DC-SIGN. A unique cooperative involvement of sugar, linker, and calixarene core is likely behind the strong avidity of DC-SIGN for these low-valent systems. We claim herein new promising candidates for the rational development of a large spectrum of antiviral therapeutics.
Collapse
Affiliation(s)
- Khouloud Chakroun
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, Amiens, 80039 Cédex, France.,Faculté des Sciences de Bizerte, Laboratoire d'Application de la Chimie aux Ressources et Substances Naturelles et à l'Environnement (LACReSNE) Unité ≪Interactions Moléculaires Spécifiques≫, Université de Carthage Zarzouna-Bizerte, Zarzouna-Bizerte, Tennessee 7021, Tunisia
| | - Marwa Taouai
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, Amiens, 80039 Cédex, France.,Faculté des Sciences de Bizerte, Laboratoire d'Application de la Chimie aux Ressources et Substances Naturelles et à l'Environnement (LACReSNE) Unité ≪Interactions Moléculaires Spécifiques≫, Université de Carthage Zarzouna-Bizerte, Zarzouna-Bizerte, Tennessee 7021, Tunisia
| | - Vanessa Porkolab
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, GrenobleF-38044, France
| | - Joanna Luczkowiak
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid 28041, Spain
| | - Roman Sommer
- Chemical Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken66123, Germany
| | - Coraline Cheneau
- Nantes Université, Inserm, CHU Nantes, Center for Research in Transplantation and Immunology UMR1064, ITUN, Nantes44093, France
| | - David Mathiron
- UFR des Sciences Bâtiment Serres-Transfert Rue Dallery, Passage du sourire d'Avril, Amiens 80039 Cedex 1, France
| | - Mohamed Amine Ben Maaouia
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, Amiens, 80039 Cédex, France.,Faculté des Sciences de Bizerte, Laboratoire d'Application de la Chimie aux Ressources et Substances Naturelles et à l'Environnement (LACReSNE) Unité ≪Interactions Moléculaires Spécifiques≫, Université de Carthage Zarzouna-Bizerte, Zarzouna-Bizerte, Tennessee 7021, Tunisia
| | - Serge Pilard
- UFR des Sciences Bâtiment Serres-Transfert Rue Dallery, Passage du sourire d'Avril, Amiens 80039 Cedex 1, France
| | - Rym Abidi
- Faculté des Sciences de Bizerte, Laboratoire d'Application de la Chimie aux Ressources et Substances Naturelles et à l'Environnement (LACReSNE) Unité ≪Interactions Moléculaires Spécifiques≫, Université de Carthage Zarzouna-Bizerte, Zarzouna-Bizerte, Tennessee 7021, Tunisia
| | - Catherine Mullié
- Laboratoire AGIR-UR UPJV 4294, UFR de Pharmacie, Université de Picardie Jules Verne, Amiens80037, France
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, GrenobleF-38044, France
| | - Peter J Cragg
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton BN2 4GJ, U.K
| | - Franck Halary
- Nantes Université, Inserm, CHU Nantes, Center for Research in Transplantation and Immunology UMR1064, ITUN, Nantes44093, France
| | - Rafael Delgado
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid 28041, Spain
| | - Mohammed Benazza
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, Amiens, 80039 Cédex, France
| |
Collapse
|
23
|
The molecular basis for the pH-dependent calcium affinity of the pattern recognition receptor langerin. J Biol Chem 2021; 296:100718. [PMID: 33989634 PMCID: PMC8219899 DOI: 10.1016/j.jbc.2021.100718] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/12/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
The C-type lectin receptor langerin plays a vital role in the mammalian defense against invading pathogens. Langerin requires a Ca2+ cofactor, the binding affinity of which is regulated by pH. Thus, Ca2+ is bound when langerin is on the membrane but released when langerin and its pathogen substrate traffic to the acidic endosome, allowing the substrate to be degraded. The change in pH is sensed by protonation of the allosteric pH sensor histidine H294. However, the mechanism by which Ca2+ is released from the buried binding site is not clear. We studied the structural consequences of protonating H294 by molecular dynamics simulations (total simulation time: about 120 μs) and Markov models. We discovered a relay mechanism in which a proton is moved into the vicinity of the Ca2+-binding site without transferring the initial proton from H294. Protonation of H294 unlocks a conformation in which a protonated lysine side chain forms a hydrogen bond with a Ca2+-coordinating aspartic acid. This destabilizes Ca2+ in the binding pocket, which we probed by steered molecular dynamics. After Ca2+ release, the proton is likely transferred to the aspartic acid and stabilized by a dyad with a nearby glutamic acid, triggering a conformational transition and thus preventing Ca2+ rebinding. These results show how pH regulation of a buried orthosteric binding site from a solvent-exposed allosteric pH sensor can be realized by information transfer through a specific chain of conformational arrangements.
Collapse
|
24
|
Ramos-Soriano J, Rojo J. Glycodendritic structures as DC-SIGN binders to inhibit viral infections. Chem Commun (Camb) 2021; 57:5111-5126. [PMID: 33977972 DOI: 10.1039/d1cc01281a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
DC-SIGN, a lectin discovered two decades ago, plays a relevant role in innate immunity. Since its discovery, it has turned out to be a target for developing antiviral drugs based on carbohydrates due to its participation in the infection process of several pathogens. A plethora of carbohydrate multivalent systems using different scaffolds have been described to achieve this goal. Our group has made significant contributions to this field, which are revised herein.
Collapse
Affiliation(s)
- Javier Ramos-Soriano
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de La Cartuja, CSIC and Universidad de Sevilla, Américo Vespucio, 49, 41092 Sevilla, Spain.
| | - Javier Rojo
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de La Cartuja, CSIC and Universidad de Sevilla, Américo Vespucio, 49, 41092 Sevilla, Spain.
| |
Collapse
|
25
|
Thépaut M, Luczkowiak J, Vivès C, Labiod N, Bally I, Lasala F, Grimoire Y, Fenel D, Sattin S, Thielens N, Schoehn G, Bernardi A, Delgado R, Fieschi F. DC/L-SIGN recognition of spike glycoprotein promotes SARS-CoV-2 trans-infection and can be inhibited by a glycomimetic antagonist. PLoS Pathog 2021; 17:e1009576. [PMID: 34015061 PMCID: PMC8136665 DOI: 10.1371/journal.ppat.1009576] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/20/2021] [Indexed: 12/26/2022] Open
Abstract
The efficient spread of SARS-CoV-2 resulted in a unique pandemic in modern history. Despite early identification of ACE2 as the receptor for viral spike protein, much remains to be understood about the molecular events behind viral dissemination. We evaluated the contribution of C-type lectin receptors (CLRS) of antigen-presenting cells, widely present in respiratory mucosa and lung tissue. DC-SIGN, L-SIGN, Langerin and MGL bind to diverse glycans of the spike using multiple interaction areas. Using pseudovirus and cells derived from monocytes or T-lymphocytes, we demonstrate that while virus capture by the CLRs examined does not allow direct cell infection, DC/L-SIGN, among these receptors, promote virus transfer to permissive ACE2+ Vero E6 cells. A glycomimetic compound designed against DC-SIGN, enable inhibition of this process. These data have been then confirmed using authentic SARS-CoV-2 virus and human respiratory cell lines. Thus, we described a mechanism potentiating viral spreading of infection.
Collapse
Affiliation(s)
- Michel Thépaut
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Corinne Vivès
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Nuria Labiod
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Isabelle Bally
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Fátima Lasala
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Yasmina Grimoire
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Daphna Fenel
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Sara Sattin
- Universita`degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| | - Nicole Thielens
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Guy Schoehn
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Anna Bernardi
- Universita`degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| |
Collapse
|
26
|
Alves I, Santos-Pereira B, Dalebout H, Santos S, Vicente MM, Campar A, Thepaut M, Fieschi F, Strahl S, Boyaval F, Vizcaíno R, Silva R, Holst-Bernal S, Vasconcelos C, Santos L, Wuhrer M, Marinho A, Heijs B, Pinho SS. Protein mannosylation as a diagnostic and prognostic biomarker of lupus nephritis: an unusual glycan-neoepitope in Systemic Lupus Erythematosus. Arthritis Rheumatol 2021; 73:2069-2077. [PMID: 33881228 DOI: 10.1002/art.41768] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/08/2021] [Indexed: 11/08/2022]
Abstract
OBJECTIVES Changes in protein glycosylation are a hallmark of immune-mediated diseases. Glycans are master regulators of the inflammatory response being important molecules for the discrimination between "self"/"non-self". We here explored whether lupus nephritis (LN) exhibit an altered cellular glycosylation aiming to identify a unique glycosignature that characterizes LN pathogenesis. METHODS A comprehensive tissue glycomics characterization was performed in a cohort of human biopsy-proven LN clinical samples from SLE patients. A combination of advanced tissue mass spectrometry imaging (MSI); in situ glyco-characterization and ex vivo glycophenotyping of human kidney biopsies were performed to structurally map the N-glycans repertoire in LN samples. RESULTS LN revealed a unique glycan signature characterized by an increased abundance and spatial distribution of unusual mannose-enriched glycans that are typically found in lower microorganisms; this glycosignature was specific of LN as it was not observed in other kidney diseases. Exposure of mannosylated glycans in LN was found to occur at the cell surface of kidney cells promoting an increased recognition by specific glycans-recognizing receptors, expressed by immune cells. This abnormal glyco-signature of LN was demonstrated to be due to a deficient complex N-glycosylation and a proficient O-mannosylation pathway. Moreover, levels of mannosylation detected in kidney biopsies from LN patients at diagnosis were demonstrated to predict the development of chronic kidney disease (CKD) with 93% of specificity. CONCLUSIONS Cellular mannosylation is a marker of LN, predicting the development of CKD, and thus constituting a potential glycobiomarker to be included in the diagnostic and prognostic algorithm of LN.
Collapse
Affiliation(s)
- Inês Alves
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Medical Faculty, University of Porto, Porto, Portugal
| | - Beatriz Santos-Pereira
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Faculty of Sciences, University of Porto, Porto, Portugal
| | - Hans Dalebout
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Sofia Santos
- Department of Nephrology, Porto University Centre Hospital, Porto, Portugal
| | - Manuel M Vicente
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Ana Campar
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.,Clinical Immunology Unit, Porto University Centre Hospital, Porto, Portugal
| | - Michel Thepaut
- Institut de Biologie Structurale, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Franck Fieschi
- Institut de Biologie Structurale, Univ. Grenoble Alpes, CNRS, CEA, Grenoble, France
| | - Sabine Strahl
- Centre for Organismal Studies (COS), Glycobiology, Heidelberg University, Heidelberg, Germany
| | - Fanny Boyaval
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ramon Vizcaíno
- Patholology Department, Porto University Centre Hospital, Porto, Portugal
| | - Roberto Silva
- Patholology Department, Centro Hospitalar São João, Porto, Portugal
| | - Stephanie Holst-Bernal
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Carlos Vasconcelos
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.,Clinical Immunology Unit, Porto University Centre Hospital, Porto, Portugal
| | - Lélita Santos
- Department of Internal Medicine, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - António Marinho
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal.,Clinical Immunology Unit, Porto University Centre Hospital, Porto, Portugal
| | - Bram Heijs
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Salomé S Pinho
- Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal.,Medical Faculty, University of Porto, Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| |
Collapse
|
27
|
Alam MM, Jarvis CM, Hincapie R, McKay CS, Schimer J, Sanhueza-Chavez CA, Xu K, Diehl RC, Finn MG, Kiessling LL. Glycan-Modified Virus-like Particles Evoke T Helper Type 1-like Immune Responses. ACS NANO 2021; 15:309-321. [PMID: 32790346 PMCID: PMC8249087 DOI: 10.1021/acsnano.0c03023] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Dendritic cells (DCs) are highly effective antigen-presenting cells that shape immune responses. Vaccines that deliver antigen to the DCs can harness their power. DC surface lectins recognize glycans not typically present on host tissue to facilitate antigen uptake and presentation. Vaccines that target these surface lectins should offer improved antigen delivery, but their efficacy will depend on how lectin targeting influences the T cell subtypes that result. We examined how antigen structure influences uptake and signaling from the C-type lectin DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin or CD209). Virus-like particles (VLPs) were engineered from bacteriophage Qβ to present an array of mannoside ligands. The VLPs were taken up by DCs and efficiently trafficked to endosomes. The signaling that ensued depended on the ligand displayed on the VLP: only those particles densely functionalized with an aryl mannoside, Qβ-Man540, elicited DC maturation and induced the expression of the proinflammatory cytokines characteristic of a T helper type 1 (TH1)-like immune response. This effect was traced to differential binding to DC-SIGN at the acidic pH of the endosome. Mice immunized with a VLP bearing the aryl mannoside, and a peptide antigen (Qβ-Ova-Man540) had antigen-specific responses, including the production of CD4+ T cells producing the activating cytokines interferon-γ and tumor necrosis factor-α. A TH1 response is critical for intracellular pathogens (e.g., viruses) and cancer; thus, our data highlight the value of targeting DC lectins for antigen delivery and validate the utility of DC-targeted VLPs as vaccine vehicles that induce cellular immunity.
Collapse
Affiliation(s)
- Mohammad Murshid Alam
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
| | - Cassie M. Jarvis
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
| | - Robert Hincapie
- School of Chemistry and Biochemistry and School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Craig S. McKay
- School of Chemistry and Biochemistry and School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Jiri Schimer
- School of Chemistry and Biochemistry and School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Carlos A Sanhueza-Chavez
- School of Chemistry and Biochemistry and School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
- Current address: Department of Pharmaceutical Sciences, St. John’s University, 8000 Utopia Pkwy. Queens, NY 11439, USA
| | - Ke Xu
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Roger C Diehl
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
| | - M. G. Finn
- School of Chemistry and Biochemistry and School of Biological Sciences, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332, USA
| | - Laura L. Kiessling
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, 02139 USA
- Corresponding Author: Laura L. Kiessling,
| |
Collapse
|
28
|
Di Pietro S, Bordoni V, Iacopini D, Achilli S, Pineschi M, Thépaut M, Fieschi F, Crotti P, Di Bussolo V. New lipophilic glycomimetic DC-SIGN ligands: Stereoselective synthesis and SPR-based binding inhibition assays. Bioorg Chem 2020; 107:104566. [PMID: 33387733 DOI: 10.1016/j.bioorg.2020.104566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 12/30/2022]
Abstract
The design and synthesis of efficient ligands for DC-SIGN is a topic of high interest, because this C-type lectin has been implicated in the early stages of many infection processes. DC-SIGN membrane-protein presents four carbohydrate-binding domains (CRD) that specifically recognize mannose and fucose. Therefore, antagonists of minimal disaccharide epitope Manα(1,2)Man, represent potentially interesting antibacterial and antiviral agents. In the recent past, we were able to develop efficient antagonists, mimics of the natural moiety, characterized by the presence of a real d-carbamannose unit which confers greater stability to enzymatic breakdown than the corresponding natural disaccharide ligand. Herein, we present the challenging stereoselective synthesis of four new amino or azide glycomimetic DC-SIGN antagonists with attractive orthogonal lipophilic substituents in C(3), C(4) or C(6) positions of the real carba unit, which were expected to establish crucial interactions with lipophilic areas of DC-SIGN CRD. The activity of the new ligands was evaluated by SPR binding inhibition assays. The interesting results obtained, allow to acquire important information about the influence of the lipophilic substituents present in specific positions of the carba scaffold. Furthermore, C(6) benzyl C(4) tosylamide pseudodisaccharide displayed a good affinity for DC-SIGN with a more favorable IC50 value than those of the previously described real carba-analogues. This study provides valuable knowledge for the implementation of further structural modifications towards improved inhibitors.
Collapse
Affiliation(s)
- Sebastiano Di Pietro
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy.
| | - Vittorio Bordoni
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | - Dalila Iacopini
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, 56125 Pisa, Italy
| | - Silvia Achilli
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Mauro Pineschi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | - Michel Thépaut
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38000 Grenoble, France
| | - Paolo Crotti
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 33, 56126 Pisa, Italy
| | - Valeria Di Bussolo
- Dipartimento di Chimica e Chimica Industriale, Università di Pisa, Via Moruzzi 13, 56125 Pisa, Italy.
| |
Collapse
|
29
|
Valverde P, Martínez JD, Cañada FJ, Ardá A, Jiménez-Barbero J. Molecular Recognition in C-Type Lectins: The Cases of DC-SIGN, Langerin, MGL, and L-Sectin. Chembiochem 2020; 21:2999-3025. [PMID: 32426893 PMCID: PMC7276794 DOI: 10.1002/cbic.202000238] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Carbohydrates play a pivotal role in intercellular communication processes. In particular, glycan antigens are key for sustaining homeostasis, helping leukocytes to distinguish damaged tissues and invading pathogens from healthy tissues. From a structural perspective, this cross-talk is fairly complex, and multiple membrane proteins guide these recognition processes, including lectins and Toll-like receptors. Since the beginning of this century, lectins have become potential targets for therapeutics for controlling and/or avoiding the progression of pathologies derived from an incorrect immune outcome, including infectious processes, cancer, or autoimmune diseases. Therefore, a detailed knowledge of these receptors is mandatory for the development of specific treatments. In this review, we summarize the current knowledge about four key C-type lectins whose importance has been steadily growing in recent years, focusing in particular on how glycan recognition takes place at the molecular level, but also looking at recent progresses in the quest for therapeutics.
Collapse
Affiliation(s)
- Pablo Valverde
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - J Daniel Martínez
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - F Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Avda Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, UPV-EHU, 48940, Leioa, Spain
| |
Collapse
|
30
|
Yen H, Liko I, Gault J, Wu D, Struwe WB, Robinson CV. Correlating Glycoforms of DC-SIGN with Stability Using a Combination of Enzymatic Digestion and Ion Mobility Mass Spectrometry. Angew Chem Int Ed Engl 2020; 59:15560-15564. [PMID: 33462887 PMCID: PMC7496578 DOI: 10.1002/anie.202005727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/05/2020] [Indexed: 01/03/2023]
Abstract
The immune scavenger protein DC-SIGN interacts with glycosylated proteins and has a putative role in facilitating viral infection. How these recognition events take place with different viruses is not clear and the effects of glycosylation on the folding and stability of DC-SIGN have not been reported. Herein, we report the development and application of a mass-spectrometry-based approach to both uncover and characterise the effects of O-glycans on the stability of DC-SIGN. We first quantify the Core 1 and 2 O-glycan structures on the carbohydrate recognition and extracellular domains of the protein using sequential exoglycosidase sequencing. Using ion mobility mass spectrometry, we show how specific O-glycans, and/or single monosaccharide substitutions, alter both the overall collision cross section and the gas-phase stability of the DC-SIGN isoforms. We find that rather than the mass or length of glycoprotein modifications, the stability of DC-SIGN is better correlated with the number of glycosylation sites.
Collapse
Affiliation(s)
- Hsin‐Yung Yen
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
- Present address: OMass TherapeuticsThe Schrodinger Building, Oxford Science ParkOxfordOX4 4GEUK
| | - Idlir Liko
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
- Present address: OMass TherapeuticsThe Schrodinger Building, Oxford Science ParkOxfordOX4 4GEUK
| | - Joseph Gault
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
| | - Di Wu
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
| | - Weston B. Struwe
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
| | - Carol V. Robinson
- Physical and Theoretical ChemistryDepartment of ChemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3TAUK
| |
Collapse
|
31
|
Achilli S, Monteiro JT, Serna S, Mayer-Lambertz S, Thépaut M, Le Roy A, Ebel C, Reichardt NC, Lepenies B, Fieschi F, Vivès C. TETRALEC, Artificial Tetrameric Lectins: A Tool to Screen Ligand and Pathogen Interactions. Int J Mol Sci 2020; 21:E5290. [PMID: 32722514 PMCID: PMC7432041 DOI: 10.3390/ijms21155290] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 12/15/2022] Open
Abstract
C-type lectin receptor (CLR)/carbohydrate recognition occurs through low affinity interactions. Nature compensates that weakness by multivalent display of the lectin carbohydrate recognition domain (CRD) at the cell surface. Mimicking these low affinity interactions in vitro is essential to better understand CLR/glycan interactions. Here, we present a strategy to create a generic construct with a tetrameric presentation of the CRD for any CLR, termed TETRALEC. We applied our strategy to a naturally occurring tetrameric CRD, DC-SIGNR, and compared the TETRALEC ligand binding capacity by synthetic N- and O-glycans microarray using three different DC-SIGNR constructs i) its natural tetrameric counterpart, ii) the monomeric CRD and iii) a dimeric Fc-CRD fusion. DC-SIGNR TETRALEC construct showed a similar binding profile to that of its natural tetrameric counterpart. However, differences observed in recognition of low affinity ligands underlined the importance of the CRD spatial arrangement. Moreover, we further extended the applications of DC-SIGNR TETRALEC to evaluate CLR/pathogens interactions. This construct was able to recognize heat-killed Candida albicans by flow cytometry and confocal microscopy, a so far unreported specificity of DC-SIGNR. In summary, the newly developed DC-SIGNR TETRALEC tool proved to be useful to unravel novel CLR/glycan interactions, an approach which could be applied to other CLRs.
Collapse
Affiliation(s)
- Silvia Achilli
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| | - João T. Monteiro
- Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (J.T.M.); (S.M.-L.); (B.L.)
| | - Sonia Serna
- Glycotechnology Laboratory, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), CIBER-BBN, Paseo Miramón 182, 20014 San Sebastian, Spain; (S.S.); (N.-C.R.)
| | - Sabine Mayer-Lambertz
- Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (J.T.M.); (S.M.-L.); (B.L.)
| | - Michel Thépaut
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| | - Aline Le Roy
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| | - Christine Ebel
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| | - Niels-Christian Reichardt
- Glycotechnology Laboratory, Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), CIBER-BBN, Paseo Miramón 182, 20014 San Sebastian, Spain; (S.S.); (N.-C.R.)
| | - Bernd Lepenies
- Immunology Unit & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (J.T.M.); (S.M.-L.); (B.L.)
| | - Franck Fieschi
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| | - Corinne Vivès
- Institut de Biologie Structurale, CEA, CNRS, University of Grenoble Alpes, F-38000 Grenoble, France; (S.A.); (M.T.); (A.L.R.); (C.E.); (F.F.)
| |
Collapse
|
32
|
Yen H, Liko I, Gault J, Wu D, Struwe WB, Robinson CV. Correlating Glycoforms of DC‐SIGN with Stability Using a Combination of Enzymatic Digestion and Ion Mobility Mass Spectrometry. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202005727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Hsin‐Yung Yen
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
- Present address: OMass Therapeutics The Schrodinger Building, Oxford Science Park Oxford OX4 4GE UK
| | - Idlir Liko
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
- Present address: OMass Therapeutics The Schrodinger Building, Oxford Science Park Oxford OX4 4GE UK
| | - Joseph Gault
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
| | - Di Wu
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
| | - Weston B. Struwe
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
| | - Carol V. Robinson
- Physical and Theoretical Chemistry Department of Chemistry University of Oxford South Parks Road Oxford OX1 3TA UK
| |
Collapse
|
33
|
de la Cruz N, Ramos-Soriano J, Reina JJ, de Paz JL, Thépaut M, Fieschi F, Sousa-Herves A, Rojo J. Influence of the reducing-end anomeric configuration of the Man9 epitope on DC-SIGN recognition. Org Biomol Chem 2020; 18:6086-6094. [DOI: 10.1039/d0ob01380c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The anomeric configuration of the reducing end of Man9 does not influence the binding to DC-SIGN.
Collapse
Affiliation(s)
- Noelia de la Cruz
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| | - Javier Ramos-Soriano
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| | - José J. Reina
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| | - José L. de Paz
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| | - Michel Thépaut
- Univ. Grenoble Alpes
- CNRS
- CEA
- Institut de Biologie Structurale
- 38000 Grenoble
| | - Franck Fieschi
- Univ. Grenoble Alpes
- CNRS
- CEA
- Institut de Biologie Structurale
- 38000 Grenoble
| | - Ana Sousa-Herves
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| | - Javier Rojo
- Glycosystems Laboratory
- Instituto de Investigaciones Químicas (IIQ)
- CSIC – Universidad de Sevilla
- Seville 41092
- Spain
| |
Collapse
|
34
|
Porkolab V, Pifferi C, Sutkeviciute I, Ordanini S, Taouai M, Thépaut M, Vivès C, Benazza M, Bernardi A, Renaudet O, Fieschi F. Development of C-type lectin-oriented surfaces for high avidity glycoconjugates: towards mimicking multivalent interactions on the cell surface. Org Biomol Chem 2020; 18:4763-4772. [DOI: 10.1039/d0ob00781a] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Here we described C-type lectin-oriented surfaces for SPR analysis. They allow the preservation of receptor topology, accessibility of binding sites, better evaluation of high avidity compounds and assessment of multivalent effect at cell surface.
Collapse
|
35
|
Keller BG, Rademacher C. Allostery in C-type lectins. Curr Opin Struct Biol 2019; 62:31-38. [PMID: 31838280 DOI: 10.1016/j.sbi.2019.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/30/2019] [Accepted: 11/04/2019] [Indexed: 10/25/2022]
Abstract
C-type lectins are the largest and most diverse family of mammalian carbohydrate-binding proteins. They share a common protein fold, which provides the unifying basis for calcium-mediated carbohydrate recognition. Their involvement in a multitude of biological functions is remarkable. Here, we review the variety of tasks these lectins are involved in alongside with the structural demands on the overall protein architecture. Subtle changes of the protein structure are implemented to cope with such diverse functional requirements. The presence of a high level of structural dynamics over a broad palette of time scales is paired with the presence of secondary binding sites and allosteric coordination of remote sites and renders this lectin fold a highly adaptable scaffold.
Collapse
Affiliation(s)
- Bettina G Keller
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Christoph Rademacher
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany; Max Planck Institute of Colloids and Interfaces, Department of Biomolecular Systems, 14424 Potsdam, Germany.
| |
Collapse
|
36
|
Rogers KJ, Brunton B, Mallinger L, Bohan D, Sevcik KM, Chen J, Ruggio N, Maury W. IL-4/IL-13 polarization of macrophages enhances Ebola virus glycoprotein-dependent infection. PLoS Negl Trop Dis 2019; 13:e0007819. [PMID: 31825972 PMCID: PMC6905523 DOI: 10.1371/journal.pntd.0007819] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/30/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ebolavirus (EBOV) outbreaks, while sporadic, cause tremendous morbidity and mortality. No therapeutics or vaccines are currently licensed; however, a vaccine has shown promise in clinical trials. A critical step towards development of effective therapeutics is a better understanding of factors that govern host susceptibility to this pathogen. As macrophages are an important cell population targeted during virus replication, we explore the effect of cytokine polarization on macrophage infection. METHODS/MAIN FINDINGS We utilized a BSL2 EBOV model virus, infectious, recombinant vesicular stomatitis virus encoding EBOV glycoprotein (GP) (rVSV/EBOV GP) in place of its native glycoprotein. Macrophages polarized towards a M2-like anti-inflammatory state by combined IL-4 and IL-13 treatment were more susceptible to rVSV/EBOV GP, but not to wild-type VSV (rVSV/G), suggesting that EBOV GP-dependent entry events were enhanced by these cytokines. Examination of RNA expression of known surface receptors that bind and internalize filoviruses demonstrated that IL-4/IL-13 stimulated expression of the C-type lectin receptor DC-SIGN in human macrophages and addition of the competitive inhibitor mannan abrogated IL-4/IL-13 enhanced infection. Two murine DC-SIGN-like family members, SIGNR3 and SIGNR5, were upregulated by IL-4/IL-13 in murine macrophages, but only SIGNR3 enhanced virus infection in a mannan-inhibited manner, suggesting that murine SIGNR3 plays a similar role to human DC-SIGN. In vivo IL-4/IL-13 administration significantly increased virus-mediated mortality in a mouse model and transfer of ex vivo IL-4/IL-13-treated murine peritoneal macrophages into the peritoneal cavity of mice enhanced pathogenesis. SIGNIFICANCE These studies highlight the ability of macrophage polarization to influence EBOV GP-dependent virus replication in vivo and ex vivo, with M2a polarization upregulating cell surface receptor expression and thereby enhancing virus replication. Our findings provide an increased understanding of the host factors in macrophages governing susceptibility to filoviruses and identify novel murine receptors mediating EBOV entry.
Collapse
Affiliation(s)
- Kai J. Rogers
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Bethany Brunton
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Laura Mallinger
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Dana Bohan
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Kristina M. Sevcik
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Jing Chen
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Natalie Ruggio
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
| | - Wendy Maury
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA United States of America
- * E-mail:
| |
Collapse
|
37
|
Medve L, Achilli S, Guzman‐Caldentey J, Thépaut M, Senaldi L, Le Roy A, Sattin S, Ebel C, Vivès C, Martin‐Santamaria S, Bernardi A, Fieschi F. Enhancing Potency and Selectivity of a DC-SIGN Glycomimetic Ligand by Fragment-Based Design: Structural Basis. Chemistry 2019; 25:14659-14668. [PMID: 31469191 PMCID: PMC6899773 DOI: 10.1002/chem.201903391] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/28/2019] [Indexed: 12/22/2022]
Abstract
Chemical modification of pseudo-dimannoside ligands guided by fragment-based design allowed for the exploitation of an ammonium-binding region in the vicinity of the mannose-binding site of DC-SIGN, leading to the synthesis of a glycomimetic antagonist (compound 16) of unprecedented affinity and selectivity against the related lectin langerin. Here, the computational design of pseudo-dimannoside derivatives as DC-SIGN ligands, their synthesis, their evaluation as DC-SIGN selective antagonists, the biophysical characterization of the DC-SIGN/16 complex, and the structural basis for the ligand activity are presented. On the way to the characterization of this ligand, an unusual bridging interaction within the crystals shed light on the plasticity and potential secondary binding sites within the DC-SIGN carbohydrate recognition domain.
Collapse
Affiliation(s)
- Laura Medve
- Dipartimento di ChimicaUniversità degli Studi di Milanovia Golgi 1920133MilanoItaly
| | - Silvia Achilli
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| | - Joan Guzman‐Caldentey
- Department of Structural and Chemical Biology, Centro de Investigaciones BiologicasCIB-CSICC/Ramiro de Maeztu, 928040MadridSpain
| | - Michel Thépaut
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| | - Luca Senaldi
- Dipartimento di ChimicaUniversità degli Studi di Milanovia Golgi 1920133MilanoItaly
| | - Aline Le Roy
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| | - Sara Sattin
- Dipartimento di ChimicaUniversità degli Studi di Milanovia Golgi 1920133MilanoItaly
| | - Christine Ebel
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| | - Corinne Vivès
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| | - Sonsoles Martin‐Santamaria
- Department of Structural and Chemical Biology, Centro de Investigaciones BiologicasCIB-CSICC/Ramiro de Maeztu, 928040MadridSpain
| | - Anna Bernardi
- Dipartimento di ChimicaUniversità degli Studi di Milanovia Golgi 1920133MilanoItaly
| | - Franck Fieschi
- Université Grenoble AlpesCNRS, CEAInstitut de Biologie Structurale38044GrenobleFrance
| |
Collapse
|
38
|
Solution structure, glycan specificity and of phenol oxidase inhibitory activity of Anopheles C-type lectins CTL4 and CTLMA2. Sci Rep 2019; 9:15191. [PMID: 31645596 PMCID: PMC6811590 DOI: 10.1038/s41598-019-51353-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/27/2019] [Indexed: 01/13/2023] Open
Abstract
Malaria, the world's most devastating parasitic disease, is transmitted between humans by mosquitoes of the Anopheles genus. An. gambiae is the principal malaria vector in Sub-Saharan Africa. The C-type lectins CTL4 and CTLMA2 cooperatively influence Plasmodium infection in the malaria vector Anopheles. Here we report the purification and biochemical characterization of CTL4 and CTLMA2 from An. gambiae and An. albimanus. CTL4 and CTLMA2 are known to form a disulfide-bridged heterodimer via an N-terminal tri-cysteine CXCXC motif. We demonstrate in vitro that CTL4 and CTLMA2 intermolecular disulfide formation is promiscuous within this motif. Furthermore, CTL4 and CTLMA2 form higher oligomeric states at physiological pH. Both lectins bind specific sugars, including glycosaminoglycan motifs with β1-3/β1-4 linkages between glucose, galactose and their respective hexosamines. Small-angle x-ray scattering data supports a compact heterodimer between the CTL domains. Recombinant CTL4/CTLMA2 is found to function in vivo, reversing the enhancement of phenol oxidase activity in dsCTL4-treated mosquitoes. We propose these molecular features underline a common function for CTL4/CTLMA2 in mosquitoes, with species and strain-specific variation in degrees of activity in response to Plasmodium infection.
Collapse
|
39
|
Fucosylated inhibitors of recently identified bangle lectin from Photorhabdus asymbiotica. Sci Rep 2019; 9:14904. [PMID: 31624296 PMCID: PMC6797808 DOI: 10.1038/s41598-019-51357-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/30/2019] [Indexed: 02/06/2023] Open
Abstract
A recently described bangle lectin (PHL) from the bacterium Photorhabdus asymbiotica was identified as a mainly fucose-binding protein that could play an important role in the host-pathogen interaction and in the modulation of host immune response. Structural studies showed that PHL is a homo-dimer that contains up to seven l-fucose-specific binding sites per monomer. For these reasons, potential ligands of the PHL lectin: α-l-fucopyranosyl-containing mono-, di-, tetra-, hexa- and dodecavalent ligands were tested. Two types of polyvalent structures were investigated – calix[4]arenes and dendrimers. The shared feature of all these structures was a C-glycosidic bond instead of the more common but physiologically unstable O-glycosidic bond. The inhibition potential of the tested structures was assessed using different techniques – hemagglutination, surface plasmon resonance, isothermal titration calorimetry, and cell cross-linking. All the ligands proved to be better than free l-fucose. The most active hexavalent dendrimer exhibited affinity three orders of magnitude higher than that of standard l-fucose. To determine the binding mode of some ligands, crystal complex PHL/fucosides 2 – 4 were prepared and studied using X-ray crystallography. The electron density in complexes proved the presence of the compounds in 6 out of 7 fucose-binding sites.
Collapse
|
40
|
Li RJE, Hogervorst TP, Achilli S, Bruijns SC, Arnoldus T, Vivès C, Wong CC, Thépaut M, Meeuwenoord NJ, van den Elst H, Overkleeft HS, van der Marel GA, Filippov DV, van Vliet SJ, Fieschi F, Codée JDC, van Kooyk Y. Systematic Dual Targeting of Dendritic Cell C-Type Lectin Receptor DC-SIGN and TLR7 Using a Trifunctional Mannosylated Antigen. Front Chem 2019; 7:650. [PMID: 31637232 PMCID: PMC6787163 DOI: 10.3389/fchem.2019.00650] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/11/2019] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are important initiators of adaptive immunity, and they possess a multitude of Pattern Recognition Receptors (PRR) to generate an adequate T cell mediated immunity against invading pathogens. PRR ligands are frequently conjugated to tumor-associated antigens in a vaccination strategy to enhance the immune response toward such antigens. One of these PPRs, DC-SIGN, a member of the C-type lectin receptor (CLR) family, has been extensively targeted with Lewis structures and mannose glycans, often presented in multivalent fashion. We synthesized a library of well-defined mannosides (mono-, di-, and tri-mannosides), based on known "high mannose" structures, that we presented in a systematically increasing number of copies (n = 1, 2, 3, or 6), allowing us to simultaneously study the effect of mannoside configuration and multivalency on DC-SIGN binding via Surface Plasmon Resonance (SPR) and flow cytometry. Hexavalent presentation of the clusters showed the highest binding affinity, with the hexa-α1,2-di-mannoside being the most potent ligand. The four highest binding hexavalent mannoside structures were conjugated to a model melanoma gp100-peptide antigen and further equipped with a Toll-like receptor 7 (TLR7)-agonist as adjuvant for DC maturation, creating a trifunctional vaccine conjugate. Interestingly, DC-SIGN affinity of the mannoside clusters did not directly correlate with antigen presentation enhancing properties and the α1,2-di-mannoside cluster with the highest binding affinity in our library even hampered T cell activation. Overall, this systematic study has demonstrated that multivalent glycan presentation can improve DC-SIGN binding but enhanced binding cannot be directly translated into enhanced antigen presentation and the sole assessment of binding affinity is thus insufficient to determine further functional biological activity. Furthermore, we show that well-defined antigen conjugates combining two different PRR ligands can be generated in a modular fashion to increase the effectiveness of vaccine constructs.
Collapse
Affiliation(s)
- Rui-Jun Eveline Li
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tim P. Hogervorst
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Silvia Achilli
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Sven C. Bruijns
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Tim Arnoldus
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Corinne Vivès
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Chung C. Wong
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Michel Thépaut
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Nico J. Meeuwenoord
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Hans van den Elst
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Herman S. Overkleeft
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Gijs A. van der Marel
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Dmitri V. Filippov
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Jeroen D. C. Codée
- Department of Bio-organic Synthesis, Faculty of Science, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
41
|
Abstract
Multivalent protein-protein interactions serve central roles in many essential biological processes, ranging from cell signaling and adhesion to pathogen recognition. Uncovering the rules that govern these intricate interactions is important not only to basic biology and chemistry but also to the applied sciences where researchers are interested in developing molecules to promote or inhibit these interactions. Here we report the synthesis and application of atomically precise inorganic cluster nanomolecules consisting of an inorganic core and a covalently linked densely packed layer of saccharides. These hybrid agents are stable under biologically relevant conditions and exhibit multivalent binding capabilities, which enable us to study the complex interactions between glycosylated structures and a dendritic cell lectin receptor. Importantly, we find that subtle changes in the molecular structure lead to significant differences in the nanomolecule's protein-binding properties. Furthermore, we demonstrate an example of using these hybrid nanomolecules to effectively inhibit protein-protein interactions in a human cell line. Ultimately, this work reveals an intricate interplay between the structural design of multivalent agents and their biological activities toward protein surfaces.
Collapse
|
42
|
Chéneau C, Coulon F, Porkolab V, Fieschi F, Laurant S, Razanajaona-Doll D, Pin JJ, Borst EM, Messerle M, Bressollette-Bodin C, Halary F. Fine Mapping the Interaction Between Dendritic Cell-Specific Intercellular Adhesion Molecule (ICAM)-3-Grabbing Nonintegrin and the Cytomegalovirus Envelope Glycoprotein B. J Infect Dis 2019; 218:490-503. [PMID: 29648611 PMCID: PMC6049025 DOI: 10.1093/infdis/jiy194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/03/2018] [Indexed: 12/22/2022] Open
Abstract
Background Human cytomegalovirus (HCMV) is a leading cause of virally induced congenital disorders and morbidities in immunocompromised individuals, ie, transplant, cancer, or acquired immune deficiency syndrome patients. Human cytomegalovirus infects virtually all cell types through the envelope glycoprotein complex gH/gL/gO with or without a contribution of the pentameric gH/gL/pUL128L. Together with gH/gL, the HCMV envelope glycoprotein B (gB) contributes to the viral fusion machinery. Methods We previously showed that gB is a ligand for the C-type lectin dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) contributing to HCMV attachment to and infection of DC-SIGN-expressing cells. However, the features of the DC-SIGN/gB interaction remain unclear. To address this point, the role of glycans on gB and the consequences of mutagenesis and antibody-mediated blockades on both partners were examined in this study. Results We identified DC-SIGN amino acid residues involved in this interaction through an extensive mutagenesis study. We also showed the importance of high-mannose N-glycans decorating the asparagine residue at position 208, demonstrating that the antigenic domain 5 on gB is involved in the interaction with DC-SIGN. Finally, antibody-mediated blockades allowed us to identify DC-SIGN as a major HCMV attachment receptor on monocyte-derived dendritic cells. Conclusions Taken together, these results have permitted us to fine-map the interaction between DC-SIGN and HCMV gB.
Collapse
Affiliation(s)
- Coraline Chéneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire (CHU) Nantes, France
| | - Flora Coulon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire (CHU) Nantes, France
| | - Vanessa Porkolab
- Université Grenoble Alpes, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Institute de Biologie Structurale, Grenoble, France
| | - Franck Fieschi
- Université Grenoble Alpes, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Institute de Biologie Structurale, Grenoble, France
| | | | | | | | - Eva Maria Borst
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Céline Bressollette-Bodin
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire (CHU) Nantes, France.,Service de Virologie Clinique, CHU Hotel Dieu, Nantes, France
| | - Franck Halary
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, France.,Institut de Transplantation Urologie Néphrologie, Centre Hospitalier Universitaire (CHU) Nantes, France
| |
Collapse
|
43
|
Yang Y, Davis I, Matsui T, Rubalcava I, Liu A. Quaternary structure of α-amino-β-carboxymuconate-ϵ-semialdehyde decarboxylase (ACMSD) controls its activity. J Biol Chem 2019; 294:11609-11621. [PMID: 31189654 PMCID: PMC6663868 DOI: 10.1074/jbc.ra119.009035] [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: 04/23/2019] [Revised: 06/10/2019] [Indexed: 01/07/2023] Open
Abstract
α-Amino-β-carboxymuconate-ϵ-semialdehyde decarboxylase (ACMSD) plays an important role in l-tryptophan degradation via the kynurenine pathway. ACMSD forms a homodimer and is functionally inactive as a monomer because its catalytic assembly requires an arginine residue from a neighboring subunit. However, how the oligomeric state and self-association of ACMSD are controlled in solution remains unexplored. Here, we demonstrate that ACMSD from Pseudomonas fluorescens can self-assemble into homodimer, tetramer, and higher-order structures. Using size-exclusion chromatography coupled with small-angle X-ray scattering (SEC-SAXS) analysis, we investigated the ACMSD tetramer structure, and fitting the SAXS data with X-ray crystal structures of the monomeric component, we could generate a pseudo-atomic structure of the tetramer. This analysis revealed a tetramer model of ACMSD as a head-on dimer of dimers. We observed that the tetramer is catalytically more active than the dimer and is in equilibrium with the monomer and dimer. Substituting a critical residue of the dimer-dimer interface, His-110, altered the tetramer dissociation profile by increasing the higher-order oligomer portion in solution without changing the X-ray crystal structure. ACMSD self-association was affected by pH, ionic strength, and other electrostatic interactions. Alignment of ACMSD sequences revealed that His-110 is highly conserved in a few bacteria that utilize nitrobenzoic acid as a sole source of carbon and energy, suggesting a dedicated functional role of ACMSD's self-assembly into the tetrameric and higher-order structures. These results indicate that the dynamic oligomerization status potentially regulates ACMSD activity and that SEC-SAXS coupled with X-ray crystallography is a powerful tool for studying protein self-association.
Collapse
Affiliation(s)
- Yu Yang
- Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Ian Davis
- Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Light Source, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, California 94025
| | - Ivan Rubalcava
- Department of Chemistry, University of Texas, San Antonio, Texas 78249
| | - Aimin Liu
- Department of Chemistry, University of Texas, San Antonio, Texas 78249, To whom correspondence should be addressed. Tel.:
210-458-7062; E-mail:
| |
Collapse
|
44
|
Taouai M, Porkolab V, Chakroun K, Cheneau C, Luczkowiak J, Abidi R, Lesur D, Cragg PJ, Halary F, Delgado R, Fieschi F, Benazza M. Unprecedented Thiacalixarene Fucoclusters as Strong Inhibitors of Ebola cis-Cell Infection and HCMV-gB Glycoprotein/DC-SIGN C-type Lectin Interaction. Bioconjug Chem 2019; 30:1114-1126. [DOI: 10.1021/acs.bioconjchem.9b00066] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Marwa Taouai
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, 80039, Amiens, France
- Faculté des Sciences de Bizerte, Laboratoire d’Application de la Chimie aux Ressources et Substances Naturelles et à l’Environnement (LACReSNE) Unité “Interactions Moléculaires Spécifiques”, Université de Carthage, Zarzouna-Bizerte, TN 7021, Tunisia
| | - Vanessa Porkolab
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38044 Grenoble, France
| | - Khouloud Chakroun
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, 80039, Amiens, France
- Faculté des Sciences de Bizerte, Laboratoire d’Application de la Chimie aux Ressources et Substances Naturelles et à l’Environnement (LACReSNE) Unité “Interactions Moléculaires Spécifiques”, Université de Carthage, Zarzouna-Bizerte, TN 7021, Tunisia
| | - Coraline Cheneau
- Centre de Recherche
en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, 44093 Nantes, France
| | - Joanna Luczkowiak
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid 28041, Spain
| | - Rym Abidi
- Faculté des Sciences de Bizerte, Laboratoire d’Application de la Chimie aux Ressources et Substances Naturelles et à l’Environnement (LACReSNE) Unité “Interactions Moléculaires Spécifiques”, Université de Carthage, Zarzouna-Bizerte, TN 7021, Tunisia
| | - David Lesur
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, 80039, Amiens, France
| | - Peter J. Cragg
- School of Pharmacy and Biomolecular Science, University of Brighton, Brighton BN2 4GJ, United Kingdom
| | - Franck Halary
- Centre de Recherche
en Transplantation et Immunologie (CRTI), UMR 1064, Inserm, Université de Nantes, 44093 Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, 44093 Nantes, France
| | - Rafael Delgado
- Laboratorio de Microbiología Molecular, Instituto de Investigación Hospital 12 de Octubre (imas12), Madrid 28041, Spain
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 38044 Grenoble, France
| | - Mohammed Benazza
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A-UMR7378-CNRS), Université de Picardie Jules Verne, 10 Rue Baudelocque, 80039, Amiens, France
| |
Collapse
|
45
|
Geissner A, Reinhardt A, Rademacher C, Johannssen T, Monteiro J, Lepenies B, Thépaut M, Fieschi F, Mrázková J, Wimmerova M, Schuhmacher F, Götze S, Grünstein D, Guo X, Hahm HS, Kandasamy J, Leonori D, Martin CE, Parameswarappa SG, Pasari S, Schlegel MK, Tanaka H, Xiao G, Yang Y, Pereira CL, Anish C, Seeberger PH. Microbe-focused glycan array screening platform. Proc Natl Acad Sci U S A 2019; 116:1958-1967. [PMID: 30670663 PMCID: PMC6369816 DOI: 10.1073/pnas.1800853116] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Interactions between glycans and glycan binding proteins are essential for numerous processes in all kingdoms of life. Glycan microarrays are an excellent tool to examine protein-glycan interactions. Here, we present a microbe-focused glycan microarray platform based on oligosaccharides obtained by chemical synthesis. Glycans were generated by combining different carbohydrate synthesis approaches including automated glycan assembly, solution-phase synthesis, and chemoenzymatic methods. The current library of more than 300 glycans is as diverse as the mammalian glycan array from the Consortium for Functional Glycomics and, due to its microbial focus, highly complementary. This glycan platform is essential for the characterization of various classes of glycan binding proteins. Applications of this glycan array platform are highlighted by the characterization of innate immune receptors and bacterial virulence factors as well as the analysis of human humoral immunity to pathogenic glycans.
Collapse
Affiliation(s)
- Andreas Geissner
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Anika Reinhardt
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Christoph Rademacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Timo Johannssen
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - João Monteiro
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Bernd Lepenies
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Michel Thépaut
- Université Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France
- CNRS, Institut de Biologie Structurale, F-38044 Grenoble, France
- Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Franck Fieschi
- Université Grenoble Alpes, Institut de Biologie Structurale, F-38044 Grenoble, France
- CNRS, Institut de Biologie Structurale, F-38044 Grenoble, France
- Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Jana Mrázková
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Michaela Wimmerova
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Frank Schuhmacher
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Sebastian Götze
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Dan Grünstein
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Xiaoqiang Guo
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Heung Sik Hahm
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jeyakumar Kandasamy
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Daniele Leonori
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Christopher E Martin
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | | | - Sandip Pasari
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Mark K Schlegel
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Hidenori Tanaka
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Guozhi Xiao
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - You Yang
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Claney L Pereira
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Chakkumkal Anish
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Peter H Seeberger
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany;
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
46
|
Wen HC, Lin CH, Huang JS, Tsai CL, Chen TF, Wang SK. Selective targeting of DC-SIGN by controlling the oligomannose pattern on a polyproline tetra-helix macrocycle scaffold. Chem Commun (Camb) 2019; 55:9124-9127. [PMID: 31298664 DOI: 10.1039/c9cc03124c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
DC-SIGN and langerin receptors both bind to oligomannose but lead to opposite effects upon encountering HIV. Because selective targeting of DC-SIGN can lead to anti-viral effects, we developed a glycoconjugate, which provides over 4800-fold selectivity for DC-SIGN over langerin, by controlling the oligomannose pattern on a polyproline tetra-helix macrocycle scaffold.
Collapse
Affiliation(s)
- Hsin-Chuan Wen
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | | | | | | | | | | |
Collapse
|
47
|
Medve L, Achilli S, Serna S, Zuccotto F, Varga N, Thépaut M, Civera M, Vivès C, Fieschi F, Reichardt N, Bernardi A. On-Chip Screening of a Glycomimetic Library with C-Type Lectins Reveals Structural Features Responsible for Preferential Binding of Dectin-2 over DC-SIGN/R and Langerin. Chemistry 2018; 24:14448-14460. [PMID: 29975429 PMCID: PMC6220942 DOI: 10.1002/chem.201802577] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/03/2018] [Indexed: 12/11/2022]
Abstract
A library of mannose‐ and fucose‐based glycomimetics was synthesized and screened in a microarray format against a set of C‐type lectin receptors (CLRs) that included DC‐SIGN, DC‐SIGNR, langerin, and dectin‐2. Glycomimetic ligands able to interact with dectin‐2 were identified for the first time. Comparative analysis of binding profiles allowed their selectivity against other CLRs to be probed.
Collapse
Affiliation(s)
- Laura Medve
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133, Milano, Italy
| | - Silvia Achilli
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Sonia Serna
- Glycotechnology laboratory, CIC biomaGUNE, Paseo Miramón 182, 20014, Donostia-San Sebastián, Spain
| | | | - Norbert Varga
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133, Milano, Italy
| | - Michel Thépaut
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Monica Civera
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133, Milano, Italy
| | - Corinne Vivès
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Franck Fieschi
- Univ. Grenoble Alpes, CEA, CNRS, Institut de Biologie Structurale, 38000, Grenoble, France
| | - Niels Reichardt
- Glycotechnology laboratory, CIC biomaGUNE, Paseo Miramón 182, 20014, Donostia-San Sebastián, Spain.,CIBER-BBN, 20014, Donostia-San Sebastián, Spain
| | - Anna Bernardi
- Dipartimento di Chimica, Università degli Studi di Milano, Via Golgi 19, 20133, Milano, Italy
| |
Collapse
|
48
|
Echeverria B, Serna S, Achilli S, Vivès C, Pham J, Thépaut M, Hokke CH, Fieschi F, Reichardt NC. Chemoenzymatic Synthesis of N-glycan Positional Isomers and Evidence for Branch Selective Binding by Monoclonal Antibodies and Human C-type Lectin Receptors. ACS Chem Biol 2018; 13:2269-2279. [PMID: 29894153 DOI: 10.1021/acschembio.8b00431] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Here, we describe a strategy for the rapid preparation of pure positional isomers of complex N-glycans to complement an existing array comprising a larger number of N-glycans and smaller glycan structures. The expanded array was then employed to study context-dependent binding of structural glycan fragments by monoclonal antibodies and C-type lectins. A partial enzymatic elongation of semiprotected core structures was combined with the protecting-group-aided separation of positional isomers by preparative HPLC. This methodology, which avoids the laborious chemical differentiation of antennae, was employed for the preparation of eight biantennary N-glycans with Galβ1,4GlcNAc (LN), GalNAcβ1,4GlcNAc (LDN), and GalNAcβ1,4[Fucα1,3]GlcNAc (LDNF) motifs presented on either one or both antennae. Screening of the binding specificities of three anti-LeX monoclonal IgM antibodies raised against S. mansoni glycans and three C-type lectin receptors of the innate immune system, namely DC-SIGN, DC-SIGNR, and LSECtin, revealed a surprising context-dependent fine specificity for the recognition of the glycan motifs. Moreover, we observed a striking selection of one individual positional isomer over the other by the C-type lectins tested, underscoring the biological relevance of the structural context of glycan elements in molecular recognition.
Collapse
Affiliation(s)
- Begoña Echeverria
- CIC biomaGUNE, Glycotechnology Laboratory, Paseo Miramón 182, 20014 San Sebastian, Spain
| | - Sonia Serna
- CIC biomaGUNE, Glycotechnology Laboratory, Paseo Miramón 182, 20014 San Sebastian, Spain
| | - Silvia Achilli
- Université Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Corinne Vivès
- Université Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Julie Pham
- CIC biomaGUNE, Glycotechnology Laboratory, Paseo Miramón 182, 20014 San Sebastian, Spain
| | - Michel Thépaut
- Université Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Cornelis H. Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Franck Fieschi
- Université Grenoble Alpes, CEA, CNRS, IBS, F-38000 Grenoble, France
| | - Niels-Christian Reichardt
- CIC biomaGUNE, Glycotechnology Laboratory, Paseo Miramón 182, 20014 San Sebastian, Spain
- CIBER-BBN, Paseo Miramón 182, 20014 San Sebastian, Spain
| |
Collapse
|
49
|
Bertolotti B, Sutkeviciute I, Ambrosini M, Ribeiro-Viana R, Rojo J, Fieschi F, Dvořáková H, Kašáková M, Parkan K, Hlaváčková M, Nováková K, Moravcová J. Polyvalent C-glycomimetics based on l-fucose or d-mannose as potent DC-SIGN antagonists. Org Biomol Chem 2018; 15:3995-4004. [PMID: 28443908 DOI: 10.1039/c7ob00322f] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The C-type lectin DC-SIGN expressed on immature dendritic cells is a promising target for antiviral drug development. Previously, we have demonstrated that mono- and divalent C-glycosides based on d-manno and l-fuco configurations are promising DC-SIGN ligands. Here, we described the convergent synthesis of C-glycoside dendrimers decorated with 4, 6, 9, and 12 α-l-fucopyranosyl units and with 9 and 12 α-d-mannopyranosyl units. Their affinity against DC-SIGN was assessed by surface plasmon resonance (SPR) assays. For comparison, parent O-glycosidic dendrimers were synthesized and tested, as well. A clear increase of both affinity and multivalency effect was observed for C-glycomimetics of both types (mannose and fucose). However, when dodecavalent C-glycosidic dendrimers were compared, there was no difference in affinity regarding the sugar unit (l-fuco, IC50 17 μM; d-manno, IC50 12 μM). For the rest of glycodendrimers with l-fucose or d-mannose attached by the O- or C-glycosidic linkage, C-glycosidic dendrimers were significantly more active. These results show that in addition to the expected physiological stability, the biological activity of C-glycoside mimetics is higher in comparison to the corresponding O-glycosides and therefore these glycomimetic multivalent systems represent potentially promising candidates for targeting DC-SIGN.
Collapse
Affiliation(s)
- Benedetta Bertolotti
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Porkolab V, Chabrol E, Varga N, Ordanini S, Sutkevičiu̅tė I, Thépaut M, García-Jiménez MJ, Girard E, Nieto PM, Bernardi A, Fieschi F. Rational-Differential Design of Highly Specific Glycomimetic Ligands: Targeting DC-SIGN and Excluding Langerin Recognition. ACS Chem Biol 2018; 13:600-608. [PMID: 29272097 DOI: 10.1021/acschembio.7b00958] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
At the surface of dendritic cells, C-type lectin receptors (CLRs) allow the recognition of carbohydrate-based PAMPS or DAMPS (pathogen- or danger-associated molecular patterns, respectively) and promote immune response regulation. However, some CLRs are hijacked by viral and bacterial pathogens. Thus, the design of ligands able to target specifically one CLR, to either modulate an immune response or to inhibit a given infection mechanism, has great potential value in therapeutic design. A case study is the selective blocking of DC-SIGN, involved notably in HIV trans-infection of T lymphocytes, without interfering with langerin-mediated HIV clearance. This is a challenging task due to their overlapping carbohydrate specificity. Toward the rational design of DC-SIGN selective ligands, we performed a comparative affinity study between DC-SIGN and langerin with natural ligands. We found that GlcNAc is recognized by both CLRs; however, selective sulfation are shown to increase the selectivity in favor of langerin. With the combination of site-directed mutagenesis and X-ray structural analysis of the langerin/GlcNS6S complex, we highlighted that 6-sulfation of the carbohydrate ligand induced langerin specificity. Additionally, the K313 residue from langerin was identified as a critical feature of its binding site. Using a rational and a differential approach in the study of CLR binding sites, we designed, synthesized, and characterized a new glycomimetic, which is highly specific for DC-SIGN vs langerin. STD NMR, SPR, and ITC characterizations show that compound 7 conserved the overall binding mode of the natural disaccharide while possessing an improved affinity and a strict specificity for DC-SIGN.
Collapse
Affiliation(s)
- Vanessa Porkolab
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Eric Chabrol
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Norbert Varga
- Università degli Studi di Milano (UniMI), Dip. Chimica, via Golgi 19, 20133, Milano, Italy
| | - Stefania Ordanini
- Università degli Studi di Milano (UniMI), Dip. Chimica, via Golgi 19, 20133, Milano, Italy
| | - Ieva Sutkevičiu̅tė
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Michel Thépaut
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Maria José García-Jiménez
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de La Cartuja, CSIC and Universidad de Sevilla, Américo Vespucio, 49, 41092 Sevilla, Spain
| | - Eric Girard
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| | - Pedro M. Nieto
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de La Cartuja, CSIC and Universidad de Sevilla, Américo Vespucio, 49, 41092 Sevilla, Spain
| | - Anna Bernardi
- Università degli Studi di Milano (UniMI), Dip. Chimica, via Golgi 19, 20133, Milano, Italy
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, F-38044 Grenoble, France
| |
Collapse
|