1
|
Ray A, Simpson JD, Demir I, Gisbert VG, Gomes DB, Amadei F, Alsteens D. From viral assembly to host interaction: AFM's contributions to virology. J Virol 2025; 99:e0087324. [PMID: 39655953 PMCID: PMC11784315 DOI: 10.1128/jvi.00873-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Viruses represent a diverse pool of obligate parasites that infect virtually every known organism, as such, their study is incredibly valuable for a range of fields including public health, medicine, agriculture, and ecology, and the development of biomedical technologies. Having evolved over millions of years, each virus has a unique and often complicated biology, that must be characterized on a case-by-case basis, even between strains of the same taxon. Owing to its nanoscale spatial resolution, atomic force microscopy (AFM) represents a powerful tool for exploring virus biology, including structural features, kinetics of binding to host cell ligands, virion self-assembly, and budding behaviors. Through the availability of numerous chemistries and advances in imaging modes, AFM is able to explore the complex web of host-virus interactions and life-cycle at a single virus level, exploring features at the level of individual bonds and molecules. Due to the wide array of techniques developed and data analysis approaches available, AFM can provide information that cannot be furnished by other modalities, especially at a single virus level. Here, we highlight the unique methods and information that can be obtained through the use of AFM, demonstrating both its utility and versatility in the study of viruses. As the technology continues to rapidly evolve, AFM is likely to remain an integral part of research, providing unique and important insight into many aspects of virology.
Collapse
Affiliation(s)
- Ankita Ray
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Joshua D. Simpson
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Irem Demir
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Victor G. Gisbert
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - David B. Gomes
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - Federico Amadei
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - David Alsteens
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Du Y, Gao J, He M, Yi M, Wu J, Feng L, Zeng B, Li Y, He R, Wang Y, Qin CF, Cui Z, Wang C. Simultaneous Blockade of CD209 and CD209L by Monoclonal Antibody Does Not Provide Sufficient Protection Against Multiple Viral Infections In Vivo. Immunology 2025. [PMID: 39783143 DOI: 10.1111/imm.13889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/18/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Many virus species, including Ebola virus, Marburg virus, SARS-CoV-2, dengue virus (DENV) and Zika virus (ZIKV), exploit CD209 and CD209L as alternative or attachment receptors for viral cis- or trans-infection. Thus, CD209 and CD209L may be critical targets for the development of therapeutic monoclonal blocking antibody drugs to disrupt the infection process caused by multiple viruses. Here, we produced a human chimeric monoclonal blocking antibody that simultaneously blocks CD209 and CD209L, namely 7-H7-B1. We show that 7-H7-B1 effectively blocks multiple pseudotyped or live viral infections in vitro, including SARS-CoV, SARS-CoV-2, Ebola virus, Marburg virus, ZIKV and DENV infections. However, the 7-H7-B1 mAb does not provide favourable protection against Zaire Ebola virus or ZIKV infection in hCD209 knock-in mice in vivo. Thus, our findings indicate that although CD209 and CD209L are critical for multiple viral infections in vitro, they may play only a partial role in viral infections in vivo.
Collapse
Affiliation(s)
- Yanyun Du
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiawang Gao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Mengjiao He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Yi
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Jiaqi Wu
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Lingyun Feng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Bo Zeng
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yangyang Li
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ruirui He
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yuan Wang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences (AMMS), Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, P.R. China
- University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Chenhui Wang
- The Key Laboratory for Human Disease Gene Study of Sichuan Province, Department of Laboratory Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Sichuan Medical Laboratory Clinical Medical Research Center, Sichuan Provincial People's Hospital, Chengdu, China
- Research Unit for Blindness Prevention of the Chinese Academy of Medical Sciences, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Langer J, Hartmann L, Snyder NL. Synthesis of a multivalent α-1,2-mannobiose ligand for targeting C-type lectins. RSC Adv 2024; 14:37950-37959. [PMID: 39610811 PMCID: PMC11603336 DOI: 10.1039/d4ra06526c] [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: 09/10/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
The importance of lectins in biological processes such as pathogen recognition, cell adhesion, and cell recognition is well documented. C-Type lectins, which require calcium for binding, play an important role in the innate immune response by engaging carbohydrates presented as part of the human and pathogen glycocalyx. For example, lectins such as MBL, Dectin-2, langerin and DC-SIGN selectively recognize mannose rich (high-mannose) structures presented as part of the glycocalyx. One common sugar binding motif that is recognized by these lectins on the pathogen glycocalyx is α-1,2-mannobiose, a disaccharide that consists of two mannose units connected via a α-1,2-linkage. To study the binding of these motifs in different contexts, synthetic replicas of α-1,2-mannobiose that can be presented in a multivalent fashion mimicking their presentation on the glycocalyx are required. Here we present the synthesis of a novel α-1,2-mannobiose analog bearing an azido linker from known precursors using a split and combine approach guided by neighboring group participation. Our approach makes it possible to achieve comparatively high yields and stereoselectivities while reducing the number of steps required to prepare such structures. We also introduce, for the first time, the trivalent presentation of our α-1,2-mannobiose ligand on a precision glycomacromolecule using copper-catalyzed azide-alkyne cycloaddition (CuAAC) to create high-mannose mimetics. Such structures have the potential to serve as probes for unlocking the rules of engagement between high-mannose glycans and C-type lectins like langerin and DC-SIGN.
Collapse
Affiliation(s)
- Jannis Langer
- Institute for Organic Chemistry and Macromolecular Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1 Düsseldorf 40225 Germany
- Institute for Macromolecular Chemistry, University Freiburg Stefan-Meier-Str. 31 79104 Freiburg i.Br. Germany
| | - Laura Hartmann
- Institute for Organic Chemistry and Macromolecular Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1 Düsseldorf 40225 Germany
- Institute for Macromolecular Chemistry, University Freiburg Stefan-Meier-Str. 31 79104 Freiburg i.Br. Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College Davidson NC 28035 USA
| |
Collapse
|
4
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [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: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
5
|
Nardini E, Rodriguez E, van Kooyk Y. The tissue glycome as regulator of immune activation and tolerance mediated by C-type lectins and Siglecs. Semin Immunol 2024; 76:101913. [PMID: 39602867 DOI: 10.1016/j.smim.2024.101913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
The immune system is a complex network of highly specialized microenvironments, denominated niches, which arise from dynamic interactions between immune and parenchymal cells as well as acellular components such as structural elements and local molecular signals. A critical, yet underexplored, layer shaping these niches is the glycome, the complete repertoire of glycans and glycoconjugates produced by cells. The glycome is prevalent in the outer membrane of cells and their secreted components, and can be sensed by glycan binding receptors on immune cells. These receptors detect changes in glycosylation and consequently modulate immune cell activity, trafficking, and signalling, altering homeostasis. Tissues like the brain and the placenta are prone to accommodate tolerance, while the gut and the thymus are sensitive to inflammation. We provide here an overview of current literature that shows the impact of altered glycosylation of tissues on host immune cells and how interference in this process may lead to new diagnostics and immune therapeutics, aiming to restore the immune balance in autoimmunity and cancer.
Collapse
Affiliation(s)
- Eleonora Nardini
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Ernesto Rodriguez
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC location Vrije Universiteit Amsterdam, Molecular Cell Biology and Immunology, De Boelelaan, Amsterdam 1117, The Netherlands; Cancer Center Amsterdam, Cancer Biology and Immunology, Amsterdam, The Netherlands; Amsterdam institute for Infection and Immunity, Cancer Immunology, Amsterdam, The Netherlands.
| |
Collapse
|
6
|
Pereira LHS, Alves ADC, Lopes GFM, da Silva BF, Vieira MS, Lopes DDO, Ferreira JMS, Lara Dos Santos L. Soluble isoforms of the DC-SIGN receptor can increase the dengue virus infection in immature dendritic cells. Braz J Infect Dis 2024; 28:103873. [PMID: 39341603 PMCID: PMC11490914 DOI: 10.1016/j.bjid.2024.103873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 08/01/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
Dengue is a disease with a high-impact on public health worldwide. Many researches have focused on the cell receptors involved in its pathogenesis. The role of soluble isoforms of DC-SIGN (Dendritic Cell-Specific ICAM-3 Grabbing Non-integrin) receptor in the process of Dengue Virus (DENV) infection is not well understood. This work proposes to evaluate changes in the infection process of Immature Dendritic Cells (iDCs) by DENV in the presence of DC-SIGN recombinant soluble isoforms 8, 10, and 12. The recombinant isoforms were built by heterologous expression, the DENV-2 was multiplied in the Aedes albopictus C6/36 cells and quantified in BHK-21 cells, and the iDCs were produced from the THP-1 strain. Infection assays were performed in the presence of iDCs, DENV-2, and isoforms 8, 10, and 12 separately at 25, 50 and 100 ng/mL. The final viral load was estimated by qPCR and statistical analysis was performed by Kruskal-Wallis and ANOVA tests. The iDC profile was confirmed by increasing expression of CD11c, CD86, and CD209 surface markers and maintaining CD14 expression. Infection assays demonstrated a 23-fold increase in DENV viral load in the presence of isoforms 8 and 10 at 100 ng/mL compared to the viral control (p < 0.05), while isoform 12 did not alter the viral load. It was possible to conclude that at 100 ng/mL isoforms (8 and 10) can interact with DENV, increasing viral infection, and potentially acting as opsonins.
Collapse
Affiliation(s)
- Lailah Horácio Sales Pereira
- Universidade Federal de São João del-Rei (UFSJ), Laboratório de Biologia Molecular, Divinópolis, MG, Brazil; Universidade Federal de São João del-Rei (UFSJ), Laboratório de Microbiologia Médica, Divinópolis, MG, Brazil
| | - Amanda do Carmo Alves
- Universidade Federal de São João del-Rei (UFSJ), Laboratório de Biologia Molecular, Divinópolis, MG, Brazil
| | | | - Brenda Fernandes da Silva
- Universidade Federal de São João del-Rei (UFSJ), Laboratório de Biologia Molecular, Divinópolis, MG, Brazil
| | - Mariana Sousa Vieira
- Universidade Federal de Minas Gerais (UFMG), Laboratório de Imunoparasitologia, Belo Horizonte, MG, Brazil
| | - Débora de Oliveira Lopes
- Universidade Federal de São João del-Rei (UFSJ), Laboratório de Biologia Molecular, Divinópolis, MG, Brazil
| | | | - Luciana Lara Dos Santos
- Universidade Federal de São João del-Rei (UFSJ), Laboratório de Biologia Molecular, Divinópolis, MG, Brazil
| |
Collapse
|
7
|
Nemli DD, Jiang X, Jakob RP, Gloder LM, Schwardt O, Rabbani S, Maier T, Ernst B, Cramer J. Thermodynamics-Guided Design Reveals a Cooperative Hydrogen Bond in DC-SIGN-targeted Glycomimetics. J Med Chem 2024; 67:13813-13828. [PMID: 38771131 DOI: 10.1021/acs.jmedchem.4c00623] [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: 05/22/2024]
Abstract
Due to the shallow and hydrophilic binding sites of carbohydrate-binding proteins, the design of glycomimetics is often complicated by high desolvation costs as well as competition with solvent. Therefore, a careful optimization of interaction vectors and ligand properties is required in the design and optimization of glycomimetics. Here, we employ thermodynamics-guided design to optimize mannose-based glycomimetics targeting the human C-type lectin receptor dendritic cell-specific intercellular adhesion molecule 3 grabbing nonintegrin (DC-SIGN), a pathogenic host factor in viral infections. By exploring ligand rigidification and hydrogen bond engineering, a monovalent glycomimetic with an unprecedented affinity for DC-SIGN in the low μM range was discovered. A matched molecular pair analysis based on microcalorimetric data revealed a stereospecific hydrogen bond interaction with Glu358/Ser360 as the origin of this cooperative and enthalpically dominated interaction. This detailed insight into the binding mechanism paves the way for an improvement of monovalent glycomimetics targeting DC-SIGN.
Collapse
Affiliation(s)
- Dilara D Nemli
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Xiaohua Jiang
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Roman P Jakob
- Department Biozentrum, Structural Area Focal Biology, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| | - Laura Muñoz Gloder
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| | - Oliver Schwardt
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Said Rabbani
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Timm Maier
- Department Biozentrum, Structural Area Focal Biology, University of Basel, Spitalstrasse 41, Basel 4056, Switzerland
| | - Beat Ernst
- Department of Pharmaceutical Sciences, Group Molecular Pharmacy, Pharmazentrum, University of Basel, Klingelbergstrasse 50, Basel CH-4056, Switzerland
| | - Jonathan Cramer
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, Düsseldorf 40225, Germany
| |
Collapse
|
8
|
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
|
9
|
Iyer K, Yan Z, Ross SR. Entry inhibitors as arenavirus antivirals. Front Microbiol 2024; 15:1382953. [PMID: 38650890 PMCID: PMC11033450 DOI: 10.3389/fmicb.2024.1382953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Arenaviruses belonging to the Arenaviridae family, genus mammarenavirus, are enveloped, single-stranded RNA viruses primarily found in rodent species, that cause severe hemorrhagic fever in humans. With high mortality rates and limited treatment options, the search for effective antivirals is imperative. Current treatments, notably ribavirin and other nucleoside inhibitors, are only partially effective and have significant side effects. The high lethality and lack of treatment, coupled with the absence of vaccines for all but Junín virus, has led to the classification of these viruses as Category A pathogens by the Centers for Disease Control (CDC). This review focuses on entry inhibitors as potential therapeutics against mammarenaviruses, which include both New World and Old World arenaviruses. Various entry inhibition strategies, including small molecule inhibitors and neutralizing antibodies, have been explored through high throughput screening, genome-wide studies, and drug repurposing. Notable progress has been made in identifying molecules that target receptor binding, internalization, or fusion steps. Despite promising preclinical results, the translation of entry inhibitors to approved human therapeutics has faced challenges. Many have only been tested in in vitro or animal models, and a number of candidates showed efficacy only against specific arenaviruses, limiting their broader applicability. The widespread existence of arenaviruses in various rodent species and their potential for their zoonotic transmission also underscores the need for rapid development and deployment of successful pan-arenavirus therapeutics. The diverse pool of candidate molecules in the pipeline provides hope for the eventual discovery of a broadly effective arenavirus antiviral.
Collapse
Affiliation(s)
| | | | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois, College of Medicine, Chicago, IL, United States
| |
Collapse
|
10
|
Steffens RC, Folda P, Fendler NL, Höhn M, Bücher-Schossau K, Kempter S, Snyder NL, Hartmann L, Wagner E, Berger S. GalNAc- or Mannose-PEG-Functionalized Polyplexes Enable Effective Lectin-Mediated DNA Delivery. Bioconjug Chem 2024; 35:351-370. [PMID: 38440876 DOI: 10.1021/acs.bioconjchem.3c00546] [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: 03/06/2024]
Abstract
A cationic, dendrimer-like oligo(aminoamide) carrier with four-arm topology based on succinoyl tetraethylene pentamine and histidines, cysteines, and N-terminal azido-lysines was screened for plasmid DNA delivery on various cell lines. The incorporated azides allow modification with various shielding agents of different polyethylene glycol (PEG) lengths and/or different ligands by copper-free click reaction, either before or after polyplex formation. Prefunctionalization was found to be advantageous over postfunctionalization in terms of nanoparticle formation, stability, and efficacy. A length of 24 ethylene oxide repetition units and prefunctionalization of ≥50% of azides per carrier promoted optimal polyplex shielding. PEG shielding resulted in drastically reduced DNA transfer, which could be successfully restored by active lectin targeting via novel GalNAc or mannose ligands, enabling enhanced receptor-mediated endocytosis of the carrier system. The involvement of the asialoglycoprotein receptor (ASGPR) in the uptake of GalNAc-functionalized polyplexes was confirmed in the ASGPR-positive hepatocarcinoma cell lines HepG2 and Huh7. Mannose-modified polyplexes showed superior cellular uptake and transfection efficacy compared to unmodified and shielded polyplexes in mannose-receptor-expressing dendritic cell-like DC2.4 cells.
Collapse
Affiliation(s)
- Ricarda C Steffens
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Paul Folda
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Nikole L Fendler
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
| | - Katharina Bücher-Schossau
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Susanne Kempter
- Faculty of Physics, LMU Munich, 80539 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Nicole L Snyder
- Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany
- Institute for Macromolecular Chemistry, University Freiburg, Stefan-Meier-Str. 31, 79104 Freiburg im Breisgau, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) Munich, 81377 Munich, Germany
- Center for NanoScience (CeNS), LMU Munich, 80799 Munich, Germany
| |
Collapse
|
11
|
Nieto-Fabregat F, Marseglia A, Thépaut M, Kleman JP, Abbas M, Le Roy A, Ebel C, Maalej M, Simorre JP, Laguri C, Molinaro A, Silipo A, Fieschi F, Marchetti R. Molecular recognition of Escherichia coli R1-type core lipooligosaccharide by DC-SIGN. iScience 2024; 27:108792. [PMID: 38299112 PMCID: PMC10828809 DOI: 10.1016/j.isci.2024.108792] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
Due to their ability to recognize carbohydrate structures, lectins emerged as potential receptors for bacterial lipopolysaccharides (LPS). Despite growing interest in investigating the association between host receptor lectins and exogenous glycan ligands, the molecular mechanisms underlying bacterial recognition by human lectins are still not fully understood. We contributed to fill this gap by unveiling the molecular basis of the interaction between the lipooligosaccharide of Escherichia coli and the dendritic cell-specific intracellular adhesion molecules (ICAM)-3 grabbing non-integrin (DC-SIGN). Specifically, a combination of different techniques, including fluorescence microscopy, surface plasmon resonance, NMR spectroscopy, and computational studies, demonstrated that DC-SIGN binds to the purified deacylated R1 lipooligosaccharide mainly through the recognition of its outer core pentasaccharide, which acts as a crosslinker between two different tetrameric units of DC-SIGN. Our results contribute to a better understanding of DC-SIGN-LPS interaction and may support the development of pharmacological and immunostimulatory strategies for bacterial infections, prevention, and therapy.
Collapse
Affiliation(s)
- Ferran Nieto-Fabregat
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Angela Marseglia
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Michel Thépaut
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Jean-Philippe Kleman
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Massilia Abbas
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Aline Le Roy
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Christine Ebel
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Meriem Maalej
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Jean-Pierre Simorre
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Cedric Laguri
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
| | - Antonio Molinaro
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Alba Silipo
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| | - Franck Fieschi
- University Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, 41 Avenue des Martyrs, 38000 Grenoble, France
- Institut Universitaire de France (IUF), Paris, France
| | - Roberta Marchetti
- Department of Chemical Science, University of Naples Federico II Via Cinthia 4, 80126 Naples, Italy
| |
Collapse
|
12
|
Dey D, Hasan SS. Strategies for rapid production of crystallization quality coatomer WD40 domains. Protein Expr Purif 2023; 212:106358. [PMID: 37625737 PMCID: PMC10529451 DOI: 10.1016/j.pep.2023.106358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
The vesicular secretion of soluble cargo proteins from the endoplasmic reticulum (ER) is accompanied by the export of ER-resident membrane proteins that are co-packaged in secretory vesicles. The cytosolic coatomer protein complex I (COPI) utilizes the N-terminal WD40 domains of α-COPI and β'-COPI subunits to bind these membrane protein "clients" for ER retrieval. These "αWD40" and "β'WD40" domains are structural homologs that demonstrate distinct selectivity for client proteins. However, elucidation of the atomic-level principles of coatomer-client interactions has been challenging due to the tendency of αWD40 domain to undergo aggregation during expression and purification. Here we describe a rapid recombinant production strategy from E. coli, which substantially enhances the quality of the purified αWD40 domain. The αWD40 purification and crystallization are completed within one day, which minimizes aggregation losses and yields a 1.9 Å resolution crystal structure. We demonstrate the versatility of this strategy by applying it to purify the β'WD40 domain, which yields crystal structures in the 1.2-1.3 Å resolution range. As an alternate recombinant production system, we develop a cost-effective strategy for αWD40 production in human Expi293 cells. Finally, we suggest a roadmap to simplify these protocols further, which is of significance for the production of WD40 mutants prone to rapid aggregation. The WD40 production strategies presented here are likely to have broad applications because the WD40 domain represents one of the largest families of biomolecular interaction modules in the eukaryotic proteome and is critical for trafficking of host as well as viral proteins such as the SARS-CoV-2 spike protein.
Collapse
Affiliation(s)
- Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - S Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA; University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, Baltimore, MD, 21201, USA; Center for Biomolecular Therapeutics, University of Maryland School of Medicine, Rockville, MD, 20850, USA.
| |
Collapse
|
13
|
Wang X, Xiao G. Recent Advances in Chemical Synthesis of Structural Domains of Lipopolysaccharides from the Commensal Gut-Associated Microbiota. Chembiochem 2023; 24:e202300552. [PMID: 37731010 DOI: 10.1002/cbic.202300552] [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: 08/04/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 09/22/2023]
Abstract
Lipopolysaccharides from the commensal gut-associated microbiota are interesting biomolecules for the treatment of various inflammatory diseases. Different from pathogenic lipopolysaccharides, commensal lipopolysaccharides have distinct chemical structures and mediate beneficial homeostasis with the immune system of the host. However, the accessibility issues of homogenous and pure commensal lipopolysaccharides hampered the in-depth studies of their functions. In this concept article, we highlight the recent synthesis of lipopolysaccharides from gut-associated lymphoid-tissue-resident Alcaligenes faecalis and Bacteroides vulgatus, which hopes to inspire the more efforts devoting to these fantastic biomolecules.
Collapse
Affiliation(s)
- Xiufang Wang
- Department of Chemistry, Kunming University, 2 Puxing Road, Kunming, 650214, China
| | - 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, 132 Lanhei Road, Kunming, 650201, China
| |
Collapse
|
14
|
Clemente B, Denis M, Silveira CP, Schiavetti F, Brazzoli M, Stranges D. Straight to the point: targeted mRNA-delivery to immune cells for improved vaccine design. Front Immunol 2023; 14:1294929. [PMID: 38090568 PMCID: PMC10711611 DOI: 10.3389/fimmu.2023.1294929] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
With the deepening of our understanding of adaptive immunity at the cellular and molecular level, targeting antigens directly to immune cells has proven to be a successful strategy to develop innovative and potent vaccines. Indeed, it offers the potential to increase vaccine potency and/or modulate immune response quality while reducing off-target effects. With mRNA-vaccines establishing themselves as a versatile technology for future applications, in the last years several approaches have been explored to target nanoparticles-enabled mRNA-delivery systems to immune cells, with a focus on dendritic cells. Dendritic cells (DCs) are the most potent antigen presenting cells and key mediators of B- and T-cell immunity, and therefore considered as an ideal target for cell-specific antigen delivery. Indeed, improved potency of DC-targeted vaccines has been proved in vitro and in vivo. This review discusses the potential specific targets for immune system-directed mRNA delivery, as well as the different targeting ligand classes and delivery systems used for this purpose.
Collapse
|
15
|
Generalov E, Yakovenko L. Receptor basis of biological activity of polysaccharides. Biophys Rev 2023; 15:1209-1222. [PMID: 37975017 PMCID: PMC10643635 DOI: 10.1007/s12551-023-01102-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/19/2023] [Indexed: 11/19/2023] Open
Abstract
Polysaccharides, the most diverse forms of organic molecules in nature, exhibit a large number of different biological activities, such as immunomodulatory, radioprotective, antioxidant, regenerative, metabolic, signaling, antitumor, and anticoagulant. The reaction of cells to a polysaccharide is determined by its specific interaction with receptors present on the cell surface, the type of cells, and their condition. The effect of many polysaccharides depends non-linearly on their concentration. The same polysaccharide in different conditions can have very different effects on cells and organisms, up to the opposite; therefore, when conducting studies of the biological activity of polysaccharides, both for the purpose of developing new drugs or approaches to the treatment of patients, and in order to clarify the features of intracellular processes, information about already known research results is needed. There is a lot of scattered data on the biological activities of polysaccharides, but there are few reviews that would consider natural polysaccharides from various sources and possible molecular mechanisms of their action. The purpose of this review is to present the main results published at different times in order to facilitate the search for information necessary for conducting relevant studies.
Collapse
Affiliation(s)
- Evgenii Generalov
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| | - Leonid Yakovenko
- Faculty of Physics, M.V. Lomonosov Moscow State University, Moscow, 119991 Russia
| |
Collapse
|
16
|
Patil V, Bohara R, Krishna Kanala V, McMahon S, Pandit A. Models and approaches to comprehend and address glial inflammation following spinal cord injury. Drug Discov Today 2023; 28:103722. [PMID: 37482236 DOI: 10.1016/j.drudis.2023.103722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Spinal cord injury (SCI) culminates in chronic inflammation and glial scar formation driven by the activation of microglia and astrocytes. Current anti-inflammatory strategies to treat glial activation associated with SCI have several limitations. Existing in vitro and ex vivo models studying molecular mechanisms associated with inflammation focus only on the acute phase. However, the progression of glial cell-derived inflammation over the acute-to-chronic phases has not been assessed. Understanding this progression will help establish a framework for evaluating therapeutic strategies. Additionally, new models could be useful as high-throughput screening (HTS) platforms. This review aims to highlight currently available models and future methods that could facilitate screening of novel therapeutics for SCI.
Collapse
Affiliation(s)
- Vaibhav Patil
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Vijaya Krishna Kanala
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Siobhan McMahon
- Anatomy, School of Medicine, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
17
|
Bui D, Favell J, Kitova EN, Li Z, McCord KA, Schmidt EN, Mozaneh F, Elaish M, El-Hawiet A, St-Pierre Y, Hobman TC, Macauley MS, Mahal LK, Flynn MR, Klassen JS. Absolute Affinities from Quantitative Shotgun Glycomics Using Concentration-Independent (COIN) Native Mass Spectrometry. ACS CENTRAL SCIENCE 2023; 9:1374-1387. [PMID: 37521792 PMCID: PMC10303200 DOI: 10.1021/acscentsci.3c00294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Indexed: 08/01/2023]
Abstract
Native mass spectrometry (nMS) screening of natural glycan libraries against glycan-binding proteins (GBPs) is a powerful tool for ligand discovery. However, as the glycan concentrations are unknown, affinities cannot be measured directly from natural libraries. Here, we introduce Concentration-Independent (COIN)-nMS, which enables quantitative screening of natural glycan libraries by exploiting slow mixing of solutions inside a nanoflow electrospray ionization emitter. The affinities (Kd) of detected GBP-glycan interactions are determined, simultaneously, from nMS analysis of their time-dependent relative abundance changes. We establish the reliability of COIN-nMS using interactions between purified glycans and GBPs with known Kd values. We also demonstrate the implementation of COIN-nMS using the catch-and-release (CaR)-nMS assay for glycosylated GBPs. The COIN-CaR-nMS results obtained for plant, fungal, viral, and human lectins with natural libraries containing hundreds of N-glycans and glycopeptides highlight the assay's versatility for discovering new ligands, precisely measuring their affinities, and uncovering "fine" specificities. Notably, the COIN-CaR-nMS results clarify the sialoglycan binding properties of the SARS-CoV-2 receptor binding domain and establish the recognition of monosialylated hybrid and biantennary N-glycans. Moreover, pharmacological depletion of host complex N-glycans reduces both pseudotyped virions and SARS-CoV-2 cell entry, suggesting that complex N-glycans may serve as attachment factors.
Collapse
Affiliation(s)
- Duong
T. Bui
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - James Favell
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Elena N. Kitova
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Zhixiong Li
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Kelli A. McCord
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Edward N. Schmidt
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Fahima Mozaneh
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Mohamed Elaish
- Department
of Cell Biology, University of Alberta, Edmonton T6G 2H7, AB, Canada
- Poultry
Diseases Department, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Amr El-Hawiet
- Department
of Pharmacognosy, Faculty of Pharmacy, Alexandria
University, Alexandria 21561, Egypt
| | - Yves St-Pierre
- Institut
National de la Recherche Scientifique (INRS), INRS-Centre Armand-Frappier
Santé Biotechnologie, Laval H7 V 1B7, QC, Canada
| | - Tom C. Hobman
- Department
of Cell Biology, University of Alberta, Edmonton T6G 2H7, AB, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton T6G 2E1, AB, Canada
- Li
Ka Shing Institute of Virology, University
of Alberta, Edmonton T6G 2E1, Alberta, Canada
| | - Matthew S. Macauley
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
- Department
of Medical Microbiology and Immunology, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Lara K. Mahal
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| | - Morris R. Flynn
- Department
of Mechanical Engineering, Faculty of Engineering, University of Alberta, Edmonton T6G 1H9, Alberta, Canada
| | - John S. Klassen
- Department
of Chemistry, University of Alberta, Edmonton T6G 2G2, Alberta, Canada
| |
Collapse
|
18
|
Chayé MAM, Gasan TA, Ozir-Fazalalikhan A, Scheenstra MR, Zawistowska-Deniziak A, van Hengel ORJ, Gentenaar M, Manurung MD, Harvey MR, Codée JDC, Chiodo F, Heijke AM, Kalinowska A, van Diepen A, Hensbergen PJ, Yazdanbakhsh M, Guigas B, Hokke CH, Smits HH. Schistosoma mansoni egg-derived thioredoxin and Sm14 drive the development of IL-10 producing regulatory B cells. PLoS Negl Trop Dis 2023; 17:e0011344. [PMID: 37363916 DOI: 10.1371/journal.pntd.0011344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/02/2023] [Indexed: 06/28/2023] Open
Abstract
During chronic schistosome infections, a complex regulatory network is induced to regulate the host immune system, in which IL-10-producing regulatory B (Breg) cells play a significant role. Schistosoma mansoni soluble egg antigens (SEA) are bound and internalized by B cells and induce both human and mouse IL-10 producing Breg cells. To identify Breg-inducing proteins in SEA, we fractionated SEA by size exclusion chromatography and found 6 fractions able to induce IL-10 production by B cells (out of 18) in the high, medium and low molecular weight (MW) range. The high MW fractions were rich in heavily glycosylated molecules, including multi-fucosylated proteins. Using SEA glycoproteins purified by affinity chromatography and synthetic glycans coupled to gold nanoparticles, we investigated the role of these glycan structures in inducing IL-10 production by B cells. Then, we performed proteomics analysis on active low MW fractions and identified a number of proteins with putative immunomodulatory properties, notably thioredoxin (SmTrx1) and the fatty acid binding protein Sm14. Subsequent splenic murine B cell stimulations and hock immunizations with recombinant SmTrx1 and Sm14 showed their ability to dose-dependently induce IL-10 production by B cells both in vitro and in vivo. Identification of unique Breg cells-inducing molecules may pave the way to innovative therapeutic strategies for inflammatory and auto-immune diseases.
Collapse
Affiliation(s)
- Mathilde A M Chayé
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas A Gasan
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Maaike R Scheenstra
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna Zawistowska-Deniziak
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
- Department of Immunology, Institute of Functional Biology and Ecology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Oscar R J van Hengel
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Max Gentenaar
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mikhael D Manurung
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael R Harvey
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Jeroen D C Codée
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Fabrizio Chiodo
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
- Italian National Research Council, Institute of Biomolecular Chemistry, Pozzuoli, Italy
| | - Anouk M Heijke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alicja Kalinowska
- Witold Stefański Institute of Parasitology, Polish Academy of Sciences, Warsaw, Poland
- Museum and Institute of Zoology, Polish Academy of Sciences, Warsaw, Poland
| | - Angela van Diepen
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Paul J Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bruno Guigas
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cornelis H Hokke
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
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
|
20
|
Simpson J, Ray A, Marcon C, dos Santos Natividade R, Dorrazehi GM, Durlet K, Koehler M, Alsteens D. Single-Molecule Analysis of SARS-CoV-2 Binding to C-Type Lectin Receptors. NANO LETTERS 2023; 23:1496-1504. [PMID: 36758952 PMCID: PMC9924085 DOI: 10.1021/acs.nanolett.2c04931] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/27/2023] [Indexed: 06/18/2023]
Abstract
Despite intense scrutiny throughout the pandemic, development of efficacious drugs against SARS-CoV-2 spread remains hindered. Understanding the underlying mechanisms of viral infection is fundamental for developing novel treatments. While angiotensin converting enzyme 2 (ACE2) is accepted as the key entry receptor of the virus, other infection mechanisms exist. Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) and its counterpart DC-SIGN-related (DC-SIGNR, also known as L-SIGN) have been recognized as possessing functional roles in COVID-19 disease and binding to SARS-CoV-2 has been demonstrated previously with ensemble and qualitative techniques. Here we examine the thermodynamic and kinetic parameters of the ligand-receptor interaction between these C-type lectins and the SARS-CoV-2 S1 protein using force-distance curve-based AFM and biolayer interferometry. We evidence that the S1 receptor binding domain is likely involved in this bond formation. Further, we employed deglycosidases and examined a nonglycosylated S1 variant to confirm the significance of glycosylation in this interaction. We demonstrate that the high affinity interactions observed occur through a mechanism distinct from that of ACE2.
Collapse
Affiliation(s)
- Joshua
D. Simpson
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Ankita Ray
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Claire Marcon
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Rita dos Santos Natividade
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Gol Mohammad Dorrazehi
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Kimberly Durlet
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Melanie Koehler
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - David Alsteens
- Louvain
Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
- Walloon
Excellence in Life Sciences and Biotechnology (WELBIO), 1300 Wavre, Belgium
| |
Collapse
|
21
|
Kumar V, Turnbull WB. Targeted delivery of oligonucleotides using multivalent protein-carbohydrate interactions. Chem Soc Rev 2023; 52:1273-1287. [PMID: 36723021 PMCID: PMC9940626 DOI: 10.1039/d2cs00788f] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Indexed: 02/02/2023]
Abstract
Cell surface protein-carbohydrate interactions are essential for tissue-specific recognition and endocytosis of viruses, some bacteria and their toxins, and many glycoproteins. Often protein-carbohydrate interactions are multivalent - multiple copies of glycans bind simultaneously to multimeric receptors. Multivalency enhances both affinity and binding specificity, and is of interest for targeted delivery of drugs to specific cell types. The first such example of carbohydrate-mediated drug delivery to reach the clinic is Givosiran, a small interfering ribonucleic acid (siRNA) that is conjugated to a trivalent N-acetylgalactosamine (GalNAc) ligand. This ligand enables efficient uptake of the nucleic acid by the asialoglycoprotein receptor (ASGP-R) on hepatocytes. Synthetic multivalent ligands for ASGP-R were among the first 'cluster glycosides' developed at the birth of multivalent glycoscience around 40 years ago. In this review we trace the history of 'GalNAc targeting' from early academic studies to current pharmaceuticals and consider what other opportunities could follow the success of this delivery technology.
Collapse
Affiliation(s)
- Vajinder Kumar
- Department of Chemistry, Akal University, Talwandi Sabo, Bathinda, Punjab, India.
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
22
|
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
|
23
|
Aloor A, Aradhya R, Venugopal P, Gopalakrishnan Nair B, Suravajhala R. Glycosylation in SARS-CoV-2 variants: A path to infection and recovery. Biochem Pharmacol 2022; 206:115335. [PMID: 36328134 PMCID: PMC9621623 DOI: 10.1016/j.bcp.2022.115335] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 10/25/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022]
Abstract
Glycan is an essential molecule that controls and drives life in a precise direction. The paucity of research in glycobiology may impede the significance of its role in the pandemic guidelines. The SARS-CoV-2 spike protein is heavily glycosylated, with 22 putative N-glycosylation sites and 17 potential O-glycosylation sites discovered thus far. It is the anchor point to the host cell ACE2 receptor, TMPRSS2, and many other host proteins that can be recognized by their immune system; hence, glycosylation is considered the primary target of vaccine development. Therefore, it is essential to know how this surface glycan plays a role in viral entry, infection, transmission, antigen, antibody responses, and disease progression. Although the vaccines are developed and applied against COVID-19, the proficiency of the immunizations is not accomplished with the current mutant variations. The role of glycosylation in SARS-CoV-2 and its receptor ACE2 with respect to other putative cell glycan receptors and the significance of glycan in host cell immunity in COVID-19 are discussed in this paper. Hence, the molecular signature of the glycan in the coronavirus infection can be incorporated into the mainstream therapeutic process.
Collapse
Affiliation(s)
- Arya Aloor
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana 690525, Kerala, India.
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana 690525, Kerala, India.
| | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana 690525, Kerala, India.
| | | | - Renuka Suravajhala
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana 690525, Kerala, India.
| |
Collapse
|
24
|
Hooper J, Liu Y, Budhadev D, Ainaga DF, Hondow N, Zhou D, Guo Y. Polyvalent Glycan Quantum Dots as a Multifunctional Tool for Revealing Thermodynamic, Kinetic, and Structural Details of Multivalent Lectin-Glycan Interactions. ACS APPLIED MATERIALS & INTERFACES 2022; 14:47385-47396. [PMID: 36194567 PMCID: PMC9614721 DOI: 10.1021/acsami.2c11111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Multivalent lectin-glycan interactions (MLGIs) are widespread and vital for biology. Their binding biophysical and structural details are thus highly valuable, not only for the understanding of binding affinity and specificity mechanisms but also for guiding the design of multivalent therapeutics against specific MLGIs. However, effective techniques that can reveal all such details remain unavailable. We have recently developed polyvalent glycan quantum dots (glycan-QDs) as a new probe for MLGIs. Using a pair of closely related tetrameric viral-binding lectins, DC-SIGN and DC-SIGNR, as model examples, we have revealed and quantified their large affinity differences in glycan-QD binding are due to distinct binding modes: with simultaneous binding for DC-SIGN and cross-linking for DC-SIGNR. Herein, we further extend the capacity of the glycan-QD probes by investigating the correlation between binding mode and binding thermodynamics and kinetics and further probing a structural basis of their binding nature. We reveal that while both lectins' binding with glycan-QDs is enthalpy driven with similar binding enthalpy changes, DC-SIGN pays a lower binding entropy penalty, resulting in a higher affinity than DC-SIGNR. We then show that DC-SIGN binding gives a single second-order kon rate, whereas DC-SIGNR gives a rapid initial binding followed by a much slower secondary interaction. We further identify a structural element in DC-SIGN, absent in DC-SIGNR, that plays an important role in maintaining DC-SIGN's MLGI character. Its removal switches the binding from being enthalpically to entropically driven and gives mixed binding modes containing both simultaneous and cross-linking binding behavior, without markedly affecting the overall binding affinity and kinetics.
Collapse
Affiliation(s)
- James Hooper
- School
of Food Science & Nutrition and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Yuanyuan Liu
- School
of Chemistry 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
| | - Dario Fernandez Ainaga
- School
of Chemical and Process Engineering, 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 & Nutrition and Astbury Centre for Structural
Molecular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
25
|
Tahirbegi B, Magness AJ, Piersimoni ME, Teng X, Hooper J, Guo Y, Knöpfel T, Willison KR, Klug DR, Ying L. Toward high-throughput oligomer detection and classification for early-stage aggregation of amyloidogenic protein. Front Chem 2022; 10:967882. [PMID: 36110142 PMCID: PMC9468268 DOI: 10.3389/fchem.2022.967882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/28/2022] [Indexed: 12/01/2022] Open
Abstract
Aggregation kinetics of proteins and peptides have been studied extensively due to their significance in many human diseases, including neurodegenerative disorders, and the roles they play in some key physiological processes. However, most of these studies have been performed as bulk measurements using Thioflavin T or other fluorescence turn-on reagents as indicators of fibrillization. Such techniques are highly successful in making inferences about the nucleation and growth mechanism of fibrils, yet cannot directly measure assembly reactions at low protein concentrations which is the case for amyloid-β (Aβ) peptide under physiological conditions. In particular, the evolution from monomer to low-order oligomer in early stages of aggregation cannot be detected. Single-molecule methods allow direct access to such fundamental information. We developed a high-throughput protocol for single-molecule photobleaching experiments using an automated fluorescence microscope. Stepwise photobleaching analysis of the time profiles of individual foci allowed us to determine stoichiometry of protein oligomers and probe protein aggregation kinetics. Furthermore, we investigated the potential application of supervised machine learning with support vector machines (SVMs) as well as multilayer perceptron (MLP) artificial neural networks to classify bleaching traces into stoichiometric categories based on an ensemble of measurable quantities derivable from individual traces. Both SVM and MLP models achieved a comparable accuracy of more than 80% against simulated traces up to 19-mer, although MLP offered considerable speed advantages, thus making it suitable for application to high-throughput experimental data. We used our high-throughput method to study the aggregation of Aβ40 in the presence of metal ions and the aggregation of α-synuclein in the presence of gold nanoparticles.
Collapse
Affiliation(s)
- Bogachan Tahirbegi
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Alastair J. Magness
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | | | - Xiangyu Teng
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - James Hooper
- School of Food Science and Nutrition and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Yuan Guo
- School of Food Science and Nutrition and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, United Kingdom
| | - Thomas Knöpfel
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Keith R. Willison
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - David R. Klug
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Liming Ying
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
26
|
Beirag N, Kumar C, Madan T, Shamji MH, Bulla R, Mitchell D, Murugaiah V, Neto MM, Temperton N, Idicula-Thomas S, Varghese PM, Kishore U. Human surfactant protein D facilitates SARS-CoV-2 pseudotype binding and entry in DC-SIGN expressing cells, and downregulates spike protein induced inflammation. Front Immunol 2022; 13:960733. [PMID: 35967323 PMCID: PMC9367475 DOI: 10.3389/fimmu.2022.960733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Lung surfactant protein D (SP-D) and Dendritic cell-specific intercellular adhesion molecules-3 grabbing non-integrin (DC-SIGN) are pathogen recognising C-type lectin receptors. SP-D has a crucial immune function in detecting and clearing pulmonary pathogens; DC-SIGN is involved in facilitating dendritic cell interaction with naïve T cells to mount an anti-viral immune response. SP-D and DC-SIGN have been shown to interact with various viruses, including SARS-CoV-2, an enveloped RNA virus that causes COVID-19. A recombinant fragment of human SP-D (rfhSP-D) comprising of α-helical neck region, carbohydrate recognition domain, and eight N-terminal Gly-X-Y repeats has been shown to bind SARS-CoV-2 Spike protein and inhibit SARS-CoV-2 replication by preventing viral entry in Vero cells and HEK293T cells expressing ACE2. DC-SIGN has also been shown to act as a cell surface receptor for SARS-CoV-2 independent of ACE2. Since rfhSP-D is known to interact with SARS-CoV-2 Spike protein and DC-SIGN, this study was aimed at investigating the potential of rfhSP-D in modulating SARS-CoV-2 infection. Coincubation of rfhSP-D with Spike protein improved the Spike Protein: DC-SIGN interaction. Molecular dynamic studies revealed that rfhSP-D stabilised the interaction between DC-SIGN and Spike protein. Cell binding analysis with DC-SIGN expressing HEK 293T and THP- 1 cells and rfhSP-D treated SARS-CoV-2 Spike pseudotypes confirmed the increased binding. Furthermore, infection assays using the pseudotypes revealed their increased uptake by DC-SIGN expressing cells. The immunomodulatory effect of rfhSP-D on the DC-SIGN: Spike protein interaction on DC-SIGN expressing epithelial and macrophage-like cell lines was also assessed by measuring the mRNA expression of cytokines and chemokines. RT-qPCR analysis showed that rfhSP-D treatment downregulated the mRNA expression levels of pro-inflammatory cytokines and chemokines such as TNF-α, IFN-α, IL-1β, IL- 6, IL-8, and RANTES (as well as NF-κB) in DC-SIGN expressing cells challenged by Spike protein. Furthermore, rfhSP-D treatment was found to downregulate the mRNA levels of MHC class II in DC expressing THP-1 when compared to the untreated controls. We conclude that rfhSP-D helps stabilise the interaction between SARS- CoV-2 Spike protein and DC-SIGN and increases viral uptake by macrophages via DC-SIGN, suggesting an additional role for rfhSP-D in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Chandan Kumar
- Biomedical Informatics Centre, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, Maharashtra, India
| | - Taruna Madan
- Department of Innate Immunity, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, India
| | - Mohamed H. Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Department of National Heart and Lung Institute and NIHR Biomedical Research Centre, Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, United Kingdom
| | - Roberta Bulla
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Daniel Mitchell
- WMS - Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Martin Mayora Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent and Greenwich, United Kingdom
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, National Institute for Research in Reproductive and Child Health, ICMR, Mumbai, Maharashtra, India
| | - Praveen M. Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
- *Correspondence: Praveen M. Varghese, ; Uday Kishore,
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
- Department of Veterinary Medicine, U.A.E. University, Al Ain, United Arab Emirates
- *Correspondence: Praveen M. Varghese, ; Uday Kishore,
| |
Collapse
|
27
|
Arnold JN, Mitchell DA. Tinker, tailor, soldier, cell: the role of C-type lectins in the defense and promotion of disease. Protein Cell 2022; 14:4-16. [PMID: 36726757 PMCID: PMC9871964 DOI: 10.1093/procel/pwac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
C-type lectins (CTLs) represent a large family of soluble and membrane-bound proteins which bind calcium dependently via carbohydrate recognition domains (CRDs) to glycan residues presented on the surface of a variety of pathogens. The deconvolution of a cell's glycan code by CTLs underpins several important physiological processes in mammals such as pathogen neutralization and opsonization, leukocyte trafficking, and the inflammatory response. However, as our knowledge of CTLs has developed it has become apparent that the role of this innate immune family of proteins can be double-edged, where some pathogens have developed approaches to subvert and exploit CTL interactions to promote infection and sustain the pathological state. Equally, CTL interactions with host glycoproteins can contribute to inflammatory diseases such as arthritis and cancer whereby, in certain contexts, they exacerbate inflammation and drive malignant progression. This review discusses the 'dual agent' roles of some of the major mammalian CTLs in both resolving and promoting infection, inflammation and inflammatory disease and highlights opportunities and emerging approaches for their therapeutic modulation.
Collapse
|
28
|
Fray M, Mathiron D, Pilard S, Lesur D, Abidi R, Barhoumi-Slimi T, Cragg PJ, BENAZZA M. Heteroglycoclusters through Unprecedented Orthogonal Chemistry Based on N‐Alkylation of N‐Acylhydrazone. European J Org Chem 2022. [DOI: 10.1002/ejoc.202101537] [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)
- Marwa Fray
- LG2A: Laboratoire de Glycochimie des Antimicrobiens et des Agroressources Chemistry 10 Rue Baudelocque 80039 Amiens FRANCE
| | - David Mathiron
- UPJV: Universite de Picardie Jules Verne Analytique 80039 Amiens FRANCE
| | - Serge Pilard
- UPJV: Universite de Picardie Jules Verne Analytique 80039 Amiens FRANCE
| | - David Lesur
- LG2A: Laboratoire de Glycochimie des Antimicrobiens et des Agroressources Analytique 10 Rue Baudelocque 80039 Amiens FRANCE
| | - Rym Abidi
- University of Carthage: Universite de Carthage Chemistry Zarzouna-Bizerte, TN 7021, Tunisia TN 7021 Bizerte TUNISIA
| | - Thouraya Barhoumi-Slimi
- University of Tunis El Manar: Universite de Tunis El Manar Structural Chemistry Faculty of Sciences of Tunis 2092 Tunis TUNISIA
| | - Peter J. Cragg
- University of Brighton School of Applied Sciences BN2 4GJ Brighton UNITED KINGDOM
| | - Mohammed BENAZZA
- Laboratoire de Glycochimie des Antimicrobiens et des Agroressources (LG2A UMR7378, CNRS), Université de Picardie Jules Verne Departement of organic Chemistry 10 Rue Baudelocque 80039 Amiens FRANCE
| |
Collapse
|
29
|
Role and therapeutic implications of protein glycosylation in neuroinflammation. Trends Mol Med 2022; 28:270-289. [PMID: 35120836 DOI: 10.1016/j.molmed.2022.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 01/06/2022] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The importance of glycosylation (post-translational attachment of glycan residues to proteins) in the context of neuroinflammation is only now beginning to be understood. Although the glycome is challenging to investigate due to its complexity, this field is gaining interest because of the emergence of novel analytical methods. These investigations offer the possibility of further understanding the molecular signature of disorders with underlying neuroinflammatory cascades. In this review, we portray the clinically relevant trends in glyconeurobiology and suggest glyco-related paths that could be targeted therapeutically to decrease neuroinflammation. A combinatorial insight from glycobiology and neurology can be harnessed to better understand neuroinflammatory-related conditions to identify relevant molecular targets.
Collapse
|
30
|
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
|
31
|
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
|
32
|
Guo L, Liang Y, Li H, Zheng H, Yang Z, Chen Y, Zhao X, Li J, Li B, Shi H, Sun M, Liu L. Epigenetic glycosylation of SARS-CoV-2 impact viral infection through DC&L-SIGN receptors. iScience 2021; 24:103426. [PMID: 34786539 PMCID: PMC8582233 DOI: 10.1016/j.isci.2021.103426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 10/06/2021] [Accepted: 11/08/2021] [Indexed: 01/06/2023] Open
Abstract
Glycosylation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike glycoprotein mediates viral entry and immune escape. While glycan site is determined by viral genetic code, glycosylation is completely dependent on host cell post-translational modification. Here, by producing SARS-CoV-2 virions from various host cell lines, viruses of different origins with diverse spike protein glycan patterns were revealed. Binding affinities to C-type lectin receptors (CLRs) DC&L-SIGN differed in the different glycan pattern virions. Although none of the CLRs supported viral productive infection, viral trans&cis-infection mediated by the CLRs were substantially changed among the different virions. Specifically, trans&cis-infection of virions with a high-mannose structure (Man5GlcNAc2) at the N1098 glycan site of the spike postfusion trimer were markedly enhanced. Considering L-SIGN co-expression with ACE2 on respiratory tract cells, our work underlines viral epigenetic glycosylation in authentic viral infection and highlights the attachment co-receptor role of DC&L-SIGN in SARS-CoV-2 infection and prevention. DC&L-SIGN are SARS-CoV-2 attachment co-receptor Viral spike (S) glycoprotein undergoes epigenetic modification during infection Epigenetic glycosylation affects viral in-cis&-trans infections through DC&L-SIGN High-mannose glycan at 1098 site of postfusion S trimer is vital for viral infection
Collapse
Affiliation(s)
- Lei Guo
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.,Kunming Key Laboratory of Children Infection and Immunity, Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Medical Center for Pediatric Diseases, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, China
| | - Yan Liang
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Heng Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huiwen Zheng
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zening Yang
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yanli Chen
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xin Zhao
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jing Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Binxiang Li
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haijing Shi
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ming Sun
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Longding Liu
- Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
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
|
34
|
Willment JA. Fc-conjugated C-type lectin receptors: Tools for understanding host-pathogen interactions. Mol Microbiol 2021; 117:632-660. [PMID: 34709692 DOI: 10.1111/mmi.14837] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022]
Abstract
The use of soluble fusion proteins of pattern recognition receptors (PRRs) used in the detection of exogenous and endogenous ligands has helped resolve the roles of PRRs in the innate immune response to pathogens, how they shape the adaptive immune response, and function in maintaining homeostasis. Using the immunoglobulin (Ig) crystallizable fragment (Fc) domain as a fusion partner, the PRR fusion proteins are soluble, stable, easily purified, have increased affinity due to the Fc homodimerization properties, and consequently have been used in a wide range of applications such as flow cytometry, screening of protein and glycan arrays, and immunofluorescent microscopy. This review will predominantly focus on the recognition of pathogens by the cell membrane-expressed glycan-binding proteins of the C-type lectin receptor (CLR) subgroup of PRRs. PRRs bind to conserved pathogen-associated molecular patterns (PAMPs), such as glycans, usually located within or on the outer surface of the pathogen. Significantly, many glycans structures are identical on both host and pathogen (e.g. the Lewis (Le) X glycan), allowing the use of Fc CLR fusion proteins with known endogenous and/or exogenous ligands as tools to identify pathogen structures that are able to interact with the immune system. Screens of highly purified pathogen-derived cell wall components have enabled identification of many unique PAMP structures recognized by CLRs. This review highlights studies using Fc CLR fusion proteins, with emphasis on the PAMPs found in fungi, bacteria, viruses, and parasites. The structure and unique features of the different CLR families is presented using examples from a broad range of microbes whenever possible.
Collapse
Affiliation(s)
- Janet A Willment
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
35
|
Gao M, Li H, Ye C, Chen K, Jiang H, Yu K. Glycan Epitopes and Potential Glycoside Antagonists of DC-SIGN Involved in COVID-19: In Silico Study. Biomolecules 2021; 11:1586. [PMID: 34827585 PMCID: PMC8615854 DOI: 10.3390/biom11111586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/19/2021] [Accepted: 10/19/2021] [Indexed: 12/28/2022] Open
Abstract
Glycosylation is an important post-translational modification that affects a wide variety of physiological functions. DC-SIGN (Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin) is a protein expressed in antigen-presenting cells that recognizes a variety of glycan epitopes. Until now, the binding of DC-SIGN to SARS-CoV-2 Spike glycoprotein has been reported in various articles and is regarded to be a factor in systemic infection and cytokine storm. The mechanism of DC-SIGN recognition offers an alternative method for discovering new medication for COVID-19 treatment. Here, we discovered three potential pockets that hold different glycan epitopes by performing molecular dynamics simulations of previously reported oligosaccharides. The "EPN" motif, "NDD" motif, and Glu354 form the most critical pocket, which is known as the Core site. We proposed that the type of glycan epitopes, rather than the precise amino acid sequence, determines the recognition. Furthermore, we deduced that oligosaccharides could occupy an additional site, which adds to their higher affinity than monosaccharides. Based on our findings and previously described glycoforms on the SARS-CoV-2 Spike, we predicted the potential glycan epitopes for DC-SIGN. It suggested that glycan epitopes could be recognized at multiple sites, not just Asn234, Asn149 and Asn343. Subsequently, we found that Saikosaponin A and Liquiritin, two plant glycosides, were promising DC-SIGN antagonists in silico.
Collapse
Affiliation(s)
- Meina Gao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.G.); (K.C.); (H.J.)
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China;
| | - Hui Li
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Chenghao Ye
- Department of Chemistry, Shantou University, Shantou 515063, China;
| | - Kaixian Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.G.); (K.C.); (H.J.)
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hualiang Jiang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China; (M.G.); (K.C.); (H.J.)
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
- Shanghai Institute for Advanced Immunochemical Studies, School of Life Science and Technology, ShanghaiTech University, Shanghai 200031, China
| | - Kunqian Yu
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
36
|
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
|
37
|
Di Maio A, Cioce A, Achilli S, Thépaut M, Vivès C, Fieschi F, Rojo J, Reichardt NC. Controlled density glycodendron microarrays for studying carbohydrate-lectin interactions. Org Biomol Chem 2021; 19:7357-7362. [PMID: 34387640 DOI: 10.1039/d1ob00872b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Glycodendron microarrays with defined valency have been constructed by on-chip synthesis on hydrophobic indium tin oxide (ITO) coated glass slides and employed in lectin-carbohydrate binding studies with several plant and human lectins. Glycodendrons presenting sugar epitopes at different valencies were prepared by spotwise strain-promoted azide-alkyne cycloaddition (SPAAC) between immobilised cyclooctyne dendrons and azide functionalised glycans. The non-covalent immobilisation of dendrons on the ITO surface by hydrophobic interaction allowed us to study dendron surface density and SPAAC conversion rate by in situ MALDI-TOF MS analysis. By diluting the dendron surface density we could study how the carbohydrate-lectin interactions became exclusively dependant on the valency of the immobilised glycodendron.
Collapse
Affiliation(s)
- Antonio Di Maio
- Glycosystems Laboratory, Instituto de Investigaciones Químicas (IIQ), CSIC - Universidad de Sevilla, Av. Américo Vespucio 49, 41092 Seville, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Srivastava AD, Unione L, Bunyatov M, Gagarinov IA, Delgado S, Abrescia NGA, Ardá A, Boons GJ. Chemoenzymatic Synthesis of Complex N-Glycans of the Parasite S. mansoni to Examine the Importance of Epitope Presentation on DC-SIGN recognition. Angew Chem Int Ed Engl 2021; 60:19287-19296. [PMID: 34124805 PMCID: PMC8456914 DOI: 10.1002/anie.202105647] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/02/2021] [Indexed: 12/14/2022]
Abstract
The importance of multivalency for N-glycan-protein interactions has primarily been studied by attachment of minimal epitopes to artificial multivalent scaffold and not in the context of multi-antennary glycans. N-glycans can be modified by bisecting GlcNAc, core xylosides and fucosides, and extended N-acetyl lactosamine moieties. The impact of such modifications on glycan recognition are also not well understood. We describe here a chemoenzymatic methodology that can provide N-glycans expressed by the parasitic worm S. mansoni having unique epitopes at each antenna and containing core xyloside. NMR, computational and electron microscopy were employed to investigate recognition of the glycans by the human lectin DC-SIGN. It revealed that core xyloside does not influence terminal epitope recognition. The multi-antennary glycans bound with higher affinity to DC-SIGN compared to mono-valent counterparts, which was attributed to proximity-induced effective concentration. The multi-antennary glycans cross-linked DC-SIGN into a dense network, which likely is relevant for antigen uptake and intracellular routing.
Collapse
Affiliation(s)
- Apoorva D Srivastava
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Luca Unione
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Mehman Bunyatov
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Ivan A Gagarinov
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands
| | - Sandra Delgado
- Molecular Recognition and Host-Pathogen Interactions, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Bizkaia Technology Park, Building 800, 48162, Derio, Bizkaia, Spain
| | - Nicola G A Abrescia
- Molecular Recognition and Host-Pathogen Interactions, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Bizkaia Technology Park, Building 800, 48162, Derio, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Bizkaia, Spain
| | - Ana Ardá
- Molecular Recognition and Host-Pathogen Interactions, CIC bioGUNE, Basque Research and Technology Alliance, BRTA, Bizkaia Technology Park, Building 800, 48162, Derio, Bizkaia, Spain.,Ikerbasque, Basque Foundation for Science, 48013, Bilbao, Bizkaia, Spain
| | - Geert-Jan Boons
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG, Utrecht, The Netherlands.,Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA, 30602, USA.,Department of Chemistry, University of Georgia, Athens, GA, 30602, USA
| |
Collapse
|
39
|
Srivastava AD, Unione L, Bunyatov M, Gagarinov IA, Delgado S, Abrescia NGA, Ardá A, Boons G. Chemoenzymatic Synthesis of Complex
N
‐Glycans of the Parasite
S. mansoni
to Examine the Importance of Epitope Presentation on DC‐SIGN recognition. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202105647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Apoorva D. Srivastava
- Department of Chemical Biology and Drug Discovery Utrecht Institute for Pharmaceutical Sciences Bijvoet Center for Biomolecular Research Utrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Luca Unione
- Department of Chemical Biology and Drug Discovery Utrecht Institute for Pharmaceutical Sciences Bijvoet Center for Biomolecular Research Utrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Mehman Bunyatov
- Department of Chemical Biology and Drug Discovery Utrecht Institute for Pharmaceutical Sciences Bijvoet Center for Biomolecular Research Utrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Ivan A. Gagarinov
- Department of Chemical Biology and Drug Discovery Utrecht Institute for Pharmaceutical Sciences Bijvoet Center for Biomolecular Research Utrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
| | - Sandra Delgado
- Molecular Recognition and Host-Pathogen Interactions CIC bioGUNE, Basque Research and Technology Alliance, BRTA Bizkaia Technology Park, Building 800 48162 Derio Bizkaia Spain
| | - Nicola G. A. Abrescia
- Molecular Recognition and Host-Pathogen Interactions CIC bioGUNE, Basque Research and Technology Alliance, BRTA Bizkaia Technology Park, Building 800 48162 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science 48013 Bilbao Bizkaia Spain
| | - Ana Ardá
- Molecular Recognition and Host-Pathogen Interactions CIC bioGUNE, Basque Research and Technology Alliance, BRTA Bizkaia Technology Park, Building 800 48162 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science 48013 Bilbao Bizkaia Spain
| | - Geert‐Jan Boons
- Department of Chemical Biology and Drug Discovery Utrecht Institute for Pharmaceutical Sciences Bijvoet Center for Biomolecular Research Utrecht University Universiteitsweg 99 3584 CG Utrecht The Netherlands
- Complex Carbohydrate Research Center University of Georgia 315 Riverbend Road Athens GA 30602 USA
- Department of Chemistry University of Georgia Athens GA 30602 USA
| |
Collapse
|
40
|
Feng R, Zhu L, Heng X, Chen G, Chen H. Immune Effect Regulated by the Chain Length: Interaction between Immune Cell Surface Receptors and Synthetic Glycopolymers. ACS APPLIED MATERIALS & INTERFACES 2021; 13:36859-36867. [PMID: 34333963 DOI: 10.1021/acsami.1c08785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Glycopolymer-based drugs for immunotherapy have attracted increasing attention because the affinity between glycans and proteins plays an important role in immune responses. Previous studies indicate that the polymer chain length influences the affinity. In the studies on enhancing the immune response by glycans, it is found that both oligosaccharides and long-chain glycopolymers work well. However, there is a lack of systematic studies on the immune enhancement effect and the binding ability of oligomers and polymers to immune-related proteins. In this paper, to study the influence of the chain length, glycopolymers based on N-acetylglucosamine with different chain lengths were synthesized, and their interaction with immune-related proteins and their effect on dendritic cell maturation were evaluated. It was proved that compared with l-glycopolymers (degree of polymerization (DP) > 20), s-glycopolymers (DP < 20) showed better binding ability to the dendritic cell-specific ICAM-3-grabbing nonintegrin protein and the toll-like receptor 4 and myeloid differentiation factor 2 complex protein by quartz crystal microbalance and molecular docking simulation. When the total sugar unit amounts are equal, s-glycopolymers are proved to be superior in promoting dendritic cell maturation by detecting the expression level of CD80 and CD86 on the surface of dendritic cells. Through the combination of experimental characterization and theoretical simulation, a deep look into the interaction between immune-related proteins and glycopolymers with different chain lengths is helpful to improve the understanding of the immune-related interactions and provides a good theoretical basis for the design of new glycopolymer-based immune drugs.
Collapse
Affiliation(s)
- Ruyan Feng
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P. R. China
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, P. R. China
| | - Lijuan Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P. R. China
| | - Xingyu Heng
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P. R. China
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, P. R. China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P. R. China
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, P. R. China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, 199 Ren'ai Road, Suzhou 215123, Jiangsu, P. R. China
| |
Collapse
|
41
|
Anderluh M, Berti F, Bzducha‐Wróbel A, Chiodo F, Colombo C, Compostella F, Durlik K, Ferhati X, Holmdahl R, Jovanovic D, Kaca W, Lay L, Marinovic‐Cincovic M, Marradi M, Ozil M, Polito L, Reina‐Martin JJ, Reis CA, Sackstein R, Silipo A, Švajger U, Vaněk O, Yamamoto F, Richichi B, van Vliet SJ. Emerging glyco-based strategies to steer immune responses. FEBS J 2021; 288:4746-4772. [PMID: 33752265 PMCID: PMC8453523 DOI: 10.1111/febs.15830] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/12/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
Glycan structures are common posttranslational modifications of proteins, which serve multiple important structural roles (for instance in protein folding), but also are crucial participants in cell-cell communications and in the regulation of immune responses. Through the interaction with glycan-binding receptors, glycans are able to affect the activation status of antigen-presenting cells, leading either to induction of pro-inflammatory responses or to suppression of immunity and instigation of immune tolerance. This unique feature of glycans has attracted the interest and spurred collaborations of glyco-chemists and glyco-immunologists to develop glycan-based tools as potential therapeutic approaches in the fight against diseases such as cancer and autoimmune conditions. In this review, we highlight emerging advances in this field, and in particular, we discuss on how glycan-modified conjugates or glycoengineered cells can be employed as targeting devices to direct tumor antigens to lectin receptors on antigen-presenting cells, like dendritic cells. In addition, we address how glycan-based nanoparticles can act as delivery platforms to enhance immune responses. Finally, we discuss some of the latest developments in glycan-based therapies, including chimeric antigen receptor (CAR)-T cells to achieve targeting of tumor-associated glycan-specific epitopes, as well as the use of glycan moieties to suppress ongoing immune responses, especially in the context of autoimmunity.
Collapse
Affiliation(s)
- Marko Anderluh
- Chair of Pharmaceutical ChemistryFaculty of PharmacyUniversity of LjubljanaSlovenia
| | | | - Anna Bzducha‐Wróbel
- Department of Biotechnology and Food MicrobiologyWarsaw University of Life Sciences‐SGGWPoland
| | - Fabrizio Chiodo
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| | - Cinzia Colombo
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Federica Compostella
- Department of Medical Biotechnology and Translational MedicineUniversity of MilanItaly
| | - Katarzyna Durlik
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Xhenti Ferhati
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Rikard Holmdahl
- Division of Medical Inflammation ResearchDepartment of Medical Biochemistry and BiophysicsKarolinska InstituteStockholmSweden
| | - Dragana Jovanovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Wieslaw Kaca
- Department of Microbiology and ParasitologyJan Kochanowski UniversityKielcePoland
| | - Luigi Lay
- Department of Chemistry and CRC Materiali Polimerici (LaMPo)University of MilanItaly
| | - Milena Marinovic‐Cincovic
- Vinča Institute of Nuclear Sciences ‐ National Institute of the Republic of SerbiaUniversity of BelgradeSerbia
| | - Marco Marradi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Musa Ozil
- Department of ChemistryFaculty of Arts and SciencesRecep Tayyip Erdogan University RizeTurkey
| | | | | | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortugal
- IPATIMUP‐Institute of Molecular Pathology and ImmunologyInstituto de Ciências Biomédicas Abel SalazarUniversity of PortoPortugal
| | - Robert Sackstein
- Department of Translational Medicinethe Translational Glycobiology InstituteHerbert Wertheim College of MedicineFlorida International UniversityMiamiFLUSA
| | - Alba Silipo
- Department of Chemical SciencesUniversity of Naples Federico IIComplesso Universitario Monte Sant’AngeloNapoliItaly
| | - Urban Švajger
- Blood Transfusion Center of SloveniaLjubljanaSlovenia
| | - Ondřej Vaněk
- Department of BiochemistryFaculty of ScienceCharles UniversityPragueCzech Republic
| | - Fumiichiro Yamamoto
- Immunohematology & Glycobiology LaboratoryJosep Carreras Leukaemia Research InstituteBadalonaSpain
| | - Barbara Richichi
- Department of Chemistry ‘Ugo Schiff’University of FlorenceFlorenceItaly
| | - Sandra J. van Vliet
- Department of Molecular Cell Biology and ImmunologyCancer Center AmsterdamAmsterdam Infection and Immunity InstituteAmsterdam UMCVrije Universiteit AmsterdamNetherlands
| |
Collapse
|
42
|
Hasan SS, Dey D, Singh S, Martin M. The Structural Biology of Eastern Equine Encephalitis Virus, an Emerging Viral Threat. Pathogens 2021; 10:pathogens10080973. [PMID: 34451437 PMCID: PMC8400090 DOI: 10.3390/pathogens10080973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Alphaviruses are arboviruses that cause arthritis and encephalitis in humans. Eastern Equine Encephalitis Virus (EEEV) is a mosquito-transmitted alphavirus that is implicated in severe encephalitis in humans with high mortality. However, limited insights are available into the fundamental biology of EEEV and residue-level details of its interactions with host proteins. In recent years, outbreaks of EEEV have been reported mainly in the United States, raising concerns about public safety. This review article summarizes recent advances in the structural biology of EEEV based mainly on single-particle cryogenic electron microscopy (cryoEM) structures. Together with functional analyses of EEEV and related alphaviruses, these structural investigations provide clues to how EEEV interacts with host proteins, which may open avenues for the development of therapeutics.
Collapse
Affiliation(s)
- S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 9600 Gudelsky Drive, Rockville, MD 20850, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201, USA
- Correspondence:
| | - Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Matthew Martin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| |
Collapse
|
43
|
Schön K, Lepenies B, Goyette-Desjardins G. Impact of Protein Glycosylation on the Design of Viral Vaccines. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2021; 175:319-354. [PMID: 32935143 DOI: 10.1007/10_2020_132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Glycans play crucial roles in various biological processes such as cell proliferation, cell-cell interactions, and immune responses. Since viruses co-opt cellular biosynthetic pathways, viral glycosylation mainly depends on the host cell glycosylation machinery. Consequently, several viruses exploit the cellular glycosylation pathway to their advantage. It was shown that viral glycosylation is strongly dependent on the host system selected for virus propagation and/or protein expression. Therefore, the use of different expression systems results in various glycoforms of viral glycoproteins that may differ in functional properties. These differences clearly illustrate that the choice of the expression system can be important, as the resulting glycosylation may influence immunological properties. In this review, we will first detail protein N- and O-glycosylation pathways and the resulting glycosylation patterns; we will then discuss different aspects of viral glycosylation in pathogenesis and in vaccine development; and finally, we will elaborate on how to harness viral glycosylation in order to optimize the design of viral vaccines. To this end, we will highlight specific examples to demonstrate how glycoengineering approaches and exploitation of different expression systems could pave the way towards better self-adjuvanted glycan-based viral vaccines.
Collapse
Affiliation(s)
- Kathleen Schön
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hanover, Germany
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, Hanover, Germany
| | - Bernd Lepenies
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hanover, Germany.
| | - Guillaume Goyette-Desjardins
- Immunology Unit and Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Hanover, Germany.
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Villar J, Salazar ML, Jiménez JM, Campo MD, Manubens A, Gleisner MA, Ávalos I, Salazar-Onfray F, Salazar F, Mitchell DA, Alshahrani MY, Martínez-Pomares L, Becker MI. C-type lectin receptors MR and DC-SIGN are involved in recognition of hemocyanins, shaping their immunostimulatory effects on human dendritic cells. Eur J Immunol 2021; 51:1715-1731. [PMID: 33891704 DOI: 10.1002/eji.202149225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 02/16/2021] [Accepted: 04/09/2021] [Indexed: 12/22/2022]
Abstract
Hemocyanins are used as immunomodulators in clinical applications because they induce a strong Th1-biased cell-mediated immunity, which has beneficial effects. They are multiligand glycosylated molecules with abundant and complex mannose-rich structures. It remains unclear whether these structures influence hemocyanin-induced immunostimulatory processes in human APCs. We have previously shown that hemocyanin glycans from Concholepas concholepas (CCH), Fissurella latimarginata (FLH), and Megathura crenulata (KLH), participate in their immune recognition and immunogenicity in mice, interacting with murine C-type lectin receptors (CLRs). Here, we studied the interactions of these hemocyanins with two major mannose-binding CLRs on monocyte-derived human DCs: MR (mannose receptor) and DC-SIGN (DC-specific ICAM-3-grabbing nonintegrin). Diverse analyses showed that hemocyanins are internalized by a mannose-sensitive mechanism. This process was calcium dependent. Moreover, hemocyanins colocalized with MR and DC-SIGN, and were partly internalized through clathrin-mediated endocytosis. The hemocyanin-mediated proinflammatory cytokine response was impaired when using deglycosylated FLH and KLH compared to CCH. We further showed that hemocyanins bind to human MR and DC-SIGN in a carbohydrate-dependent manner with affinity constants in the physiological concentration range. Overall, we showed that these three clinically valuable hemocyanins interact with human mannose-sensitive CLRs, initiating an immune response and promoting a Th1 cell-driving potential.
Collapse
Affiliation(s)
- Javiera Villar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Michelle L Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - José M Jiménez
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Miguel Del Campo
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile
| | - Augusto Manubens
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Biosonda Corporation, Santiago, Chile
| | - María Alejandra Gleisner
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Ignacio Ávalos
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Universidad de Chile, Santiago, Chile
| | - Fabián Salazar
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Medical Research Council (MRC) Centre for Medical Mycology, University of Exeter, Exeter, United Kingdom
| | - Daniel A Mitchell
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | - María Inés Becker
- Fundación Ciencia y Tecnología para el Desarrollo (FUCITED), Santiago, Chile.,Biosonda Corporation, Santiago, Chile
| |
Collapse
|
47
|
Kononova S, Litvinova E, Vakhitov T, Skalinskaya M, Sitkin S. Acceptive Immunity: The Role of Fucosylated Glycans in Human Host-Microbiome Interactions. Int J Mol Sci 2021; 22:ijms22083854. [PMID: 33917768 PMCID: PMC8068183 DOI: 10.3390/ijms22083854] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
The growth in the number of chronic non-communicable diseases in the second half of the past century and in the first two decades of the new century is largely due to the disruption of the relationship between the human body and its symbiotic microbiota, and not pathogens. The interaction of the human immune system with symbionts is not accompanied by inflammation, but is a physiological norm. This is achieved via microbiota control by the immune system through a complex balance of pro-inflammatory and suppressive responses, and only a disturbance of this balance can trigger pathophysiological mechanisms. This review discusses the establishment of homeostatic relationships during immune system development and intestinal bacterial colonization through the interaction of milk glycans, mucins, and secretory immunoglobulins. In particular, the role of fucose and fucosylated glycans in the mechanism of interactions between host epithelial and immune cells is discussed.
Collapse
Affiliation(s)
- Svetlana Kononova
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Institute of Protein Research, Russian Academy of Sciences, 142290 Pushchino, Russia
- Correspondence:
| | - Ekaterina Litvinova
- Scientific-Research Institute of Neurosciences and Medicine, 630117 Novosibirsk, Russia;
- Siberian Federal Scientific Center of Agro-BioTechnologies, Russian Academy of Sciences, Krasnoobsk, 633501 Novosibirsk, Russia
| | - Timur Vakhitov
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
| | - Maria Skalinskaya
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
| | - Stanislav Sitkin
- Department of Microbiology, State Research Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.V.); (M.S.); (S.S.)
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University Named after I.I. Mechnikov, 191015 St. Petersburg, Russia
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
| |
Collapse
|
48
|
Báez Bolivar EG, Bui DT, Kitova EN, Han L, Zheng RB, Luber EJ, Sayed SY, Mahal LK, Klassen JS. Submicron Emitters Enable Reliable Quantification of Weak Protein-Glycan Interactions by ESI-MS. Anal Chem 2021; 93:4231-4239. [PMID: 33630563 DOI: 10.1021/acs.analchem.0c05003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interactions between carbohydrates (glycans) and glycan-binding proteins (GBPs) regulate a wide variety of important biological processes. However, the affinities of most monovalent glycan-GBP complexes are typically weak (dissociation constant (Kd) > μM) and difficult to reliably measure with conventional assays; consequently, the glycan specificities of most GBPs are not well established. Here, we demonstrate how electrospray ionization mass spectrometry (ESI-MS), implemented with nanoflow ESI emitters with inner diameters of ∼50 nm, allows for the facile quantification of low-affinity glycan-GBP interactions. The small size of the droplets produced from these submicron emitters effectively eliminates the formation of nonspecific glycan-GBP binding (false positives) during the ESI process up to ∼mM glycan concentrations. Thus, interactions with affinities as low as ∼5 mM can be measured directly from the mass spectrum. The general suppression of nonspecific adducts (including nonvolatile buffers and salts) achieved with these tips enables ESI-MS glycan affinity measurements to be performed on C-type lectins, a class of GBPs that bind glycans in a calcium-dependent manner and are important regulators of immune response. At physiologically relevant calcium ion concentrations (2-3 mM), the extent of Ca2+ nonspecific adduct formation observed using the submicron emitters is dramatically suppressed, allowing glycan affinities, and the influence of Ca2+ thereon, to be measured. Finally, we show how the use of submicron emitters and suppression of nonspecific binding enable the quantification of labile (prone to in-source dissociation) glycan-GBP interactions.
Collapse
Affiliation(s)
- Erick G Báez Bolivar
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Duong T Bui
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Elena N Kitova
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Ling Han
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Ruixiang B Zheng
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Erik J Luber
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Sayed Youssef Sayed
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada T6G 2G2
| |
Collapse
|
49
|
Identification of Important N-Linked Glycosylation Sites in the Hemagglutinin Protein and Their Functional Impact on DC-SIGN Mediated Avian Influenza H5N1 Infection. Int J Mol Sci 2021; 22:ijms22020743. [PMID: 33451024 PMCID: PMC7828482 DOI: 10.3390/ijms22020743] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022] Open
Abstract
DC-SIGN, a C-type lectin mainly expressed in dendritic cells (DCs), has been reported to mediate several viral infections. We previously reported that DC-SIGN mediated H5N1 influenza A virus (AIVs) infection, however, the important DC-SIGN interaction with N-glycosylation sites remain unknown. This study aims to identify the optimal DC-SIGN interacting N-glycosylation sites in HA proteins of H5N1-AIVs. Results from NetNGlyc program analyzed the H5 hemagglutinin sequences of isolates during 2004–2020, revealing that seven and two conserved N-glycosylation sites were detected in HA1 and HA2 domain, respectively. A lentivirus pseudotyped A/Vietnam/1203/04 H5N1 envelope (H5N1-PVs) was generated which displayed an abundance of HA5 proteins on the virions via immuno-electron microscope observation. Further, H5N1-PVs or reverse-genetics (H5N1-RG) strains carrying a serial N-glycosylated mutation was generated by site-directed mutagenesis assay. Human recombinant DC-SIGN (rDC-SIGN) coated ELISA showed that H5N1-PVs bound to DC-SIGN, however, mutation on the N27Q, N39Q, and N181Q significantly reduced this binding (p < 0.05). Infectivity and capture assay demonstrated that N27Q and N39Q mutations significantly ameliorated DC-SIGN mediated H5N1 infection. Furthermore, combined mutations (N27Q&N39Q) significantly waned the interaction on either H5N1-PVs or -RG infection in cis and in trans (p < 0.01). This study concludes that N27 and N39 are two essential N-glycosylation contributing to DC-SIGN mediating H5N1 infection.
Collapse
|
50
|
Sales Pereira LH, Alves ADC, Siqueira Ferreira JM, Dos Santos LL. Soluble DC-SIGN isoforms: Ligands with unknown functions - A mini-review. Microb Pathog 2021; 150:104731. [PMID: 33429051 DOI: 10.1016/j.micpath.2021.104731] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/23/2020] [Accepted: 12/27/2020] [Indexed: 12/01/2022]
Abstract
The present study aimed to perform a mini-review of the complete soluble isoforms of Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin (sDC-SIGN), their functions, and their correlation with diseases. The present review reveals the lack of studies regarding these soluble isoforms and poor understanding of the importance of the topic, considering the concordant findings on the relevant influence of sDC-SIGN in the viral and bacterial infection process, in addition to its possible use as a cancer marker.
Collapse
Affiliation(s)
- Lailah Horácio Sales Pereira
- Laboratório de Biologia Molecular, Universidade Federal de São João Del Rei (UFSJ), Divinopolis, Minas Gerais, Brazil; Laboratório de Virologia, Universidade Federal de São João Del Rei (UFSJ), Divinopolis, Minas Gerais, Brazil.
| | - Amanda do Carmo Alves
- Laboratório de Biologia Molecular, Universidade Federal de São João Del Rei (UFSJ), Divinopolis, Minas Gerais, Brazil.
| | | | - Luciana Lara Dos Santos
- Laboratório de Biologia Molecular, Universidade Federal de São João Del Rei (UFSJ), Divinopolis, Minas Gerais, Brazil.
| |
Collapse
|