1
|
Wu X, Goebbels M, Debski-Antoniak O, Marougka K, Chao L, Smits T, Wennekes T, van Kuppeveld FJM, de Vries E, de Haan CAM. Unraveling dynamics of paramyxovirus-receptor interactions using nanoparticles displaying hemagglutinin-neuraminidase. PLoS Pathog 2024; 20:e1012371. [PMID: 39052678 PMCID: PMC11302929 DOI: 10.1371/journal.ppat.1012371] [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: 03/20/2024] [Revised: 08/06/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Sialoglycan-binding enveloped viruses often possess receptor-destroying activity to avoid being immobilized by non-functional decoy receptors. Sialic acid (Sia)-binding paramyxoviruses contain a hemagglutinin-neuraminidase (HN) protein that possesses both Sia-binding and -cleavage activities. The multivalent, dynamic receptor interactions of paramyxovirus particles provide virion motility and are a key determinant of host tropism. However, such multivalent interactions have not been exhaustively analyzed, because such studies are complicated by the low affinity of the individual interactions and the requirement of high titer virus stocks. Moreover, the dynamics of multivalent particle-receptor interactions are difficult to predict from Michaelis-Menten enzyme kinetics. Therefore, we here developed Ni-NTA nanoparticles that multivalently display recombinant soluble HN tetramers via their His tags (HN-NPs). Applying this HN-NP platform to Newcastle disease virus (NDV), we investigated using biolayer interferometry (BLI) the role of important HN residues in receptor-interactions and analyzed long-range effects between the catalytic site and the second Sia binding site (2SBS). The HN-NP system was also applicable to other paramyxoviruses. Comparative analysis of HN-NPs revealed and confirmed differences in dynamic receptor-interactions between type 1 human and murine parainfluenza viruses as well as of lab-adapted and clinical isolates of human parainfluenza virus type 3, which are likely to contribute to differences in tropism of these viruses. We propose this novel platform to be applicable to elucidate the dynamics of multivalent-receptor interactions important for host tropism and pathogenesis, particularly for difficult to grow sialoglycan-binding (paramyxo)viruses.
Collapse
Affiliation(s)
- Xuesheng Wu
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Maite Goebbels
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Oliver Debski-Antoniak
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Katherine Marougka
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Lemeng Chao
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Tony Smits
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Tom Wennekes
- Department Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Frank J. M. van Kuppeveld
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Erik de Vries
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Cornelis A. M. de Haan
- Section Virology, Division Infectious Diseases and Immunology, Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
2
|
Negi G, Sharma A, Chaudhary M, Gupta D, Harshan KH, Parveen N. SARS-CoV-2 Binding to Terminal Sialic Acid of Gangliosides Embedded in Lipid Membranes. ACS Infect Dis 2023; 9:1346-1361. [PMID: 37145972 DOI: 10.1021/acsinfecdis.3c00106] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Multiple recent reports indicate that the S protein of SARS-CoV-2 specifically interacts with membrane receptors and attachment factors other than ACE2. They likely have an active role in cellular attachment and entry of the virus. In this article, we examined the binding of SARS-CoV-2 particles to gangliosides embedded in supported lipid bilayers (SLBs), mimicking the cell membrane-like environment. We show that the virus specifically binds to sialylated (sialic acid (SIA)) gangliosides, i.e., GD1a, GM3, and GM1, as determined from the acquired single-particle fluorescence images using a time-lapse total internal reflection fluorescence (TIRF) microscope. The data of virus binding events, the apparent binding rate constant, and the maximum virus coverage on the ganglioside-rich SLBs show that the virus particles have a higher binding affinity toward the GD1a and GM3 compared to the GM1 ganglioside. Enzymatic hydrolysis of the SIA-Gal bond of the gangliosides confirms that the SIA sugar unit of GD1a and GM3 is essential for virus attachment to the SLBs and even the cell surface sialic acid is critical for the cellular attachment of the virus. The structural difference between GM3/GD1a and GM1 is the presence of SIA at the main or branched chain. We conclude that the number of SIA per ganglioside can weakly influence the initial binding rate of SARS-CoV-2 particles, whereas the terminal or more exposed SIA is critical for the virus binding to the gangliosides in SLBs.
Collapse
Affiliation(s)
- Geetanjali Negi
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Anurag Sharma
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Monika Chaudhary
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| | - Divya Gupta
- CSIR-Centre for Cellular and Molecular Biology, 500007 Hyderabad, India
| | - Krishnan H Harshan
- CSIR-Centre for Cellular and Molecular Biology, 500007 Hyderabad, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Nagma Parveen
- Department of Chemistry, Indian Institute of Technology Kanpur, 208016 Kanpur, India
| |
Collapse
|
3
|
Martino RA, Wang Q, Xu H, Hu G, Bell P, Arroyo EJ, Sims JJ, Wilson JM. Vector Affinity and Receptor Distribution Define Tissue-Specific Targeting in an Engineered AAV Capsid. J Virol 2023; 97:e0017423. [PMID: 37199615 PMCID: PMC10308920 DOI: 10.1128/jvi.00174-23] [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: 02/01/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Unbiased in vivo selections of diverse capsid libraries can yield engineered capsids that overcome gene therapy delivery challenges like traversing the blood-brain barrier (BBB), but little is known about the parameters of capsid-receptor interactions that govern their improved activity. This hampers broader efforts in precision capsid engineering and is a practical impediment to ensuring the translatability of capsid properties between preclinical animal models and human clinical trials. In this work, we utilize the adeno-associated virus (AAV)-PHP.B-Ly6a model system to better understand the targeted delivery and BBB penetration properties of AAV vectors. This model offers a defined capsid-receptor pair that can be used to systematically define relationships between target receptor affinity and in vivo activity of engineered AAV vectors. Here, we report a high-throughput method for quantifying capsid-receptor affinity and demonstrate that direct binding assays can be used to organize a vector library into families with varied affinity for their target receptor. Our data indicate that efficient central nervous system transduction requires high levels of target receptor expression at the BBB, but it is not a requirement for receptor expression to be limited to the target tissue. We observed that enhanced receptor affinity leads to reduced transduction of off-target tissues but can negatively impact on-target cellular transduction and penetration of endothelial barriers. Together, this work provides a set of tools for defining vector-receptor affinities and demonstrates how receptor expression and affinity interact to impact the performance of engineered AAV vectors in targeting the central nervous system. IMPORTANCE Novel methods for measuring adeno-associated virus (AAV)-receptor affinities, especially in relation to vector performance in vivo, would be useful to capsid engineers as they develop AAV vectors for gene therapy applications and characterize their interactions with native or engineered receptors. Here, we use the AAV-PHP.B-Ly6a model system to assess the impact of receptor affinity on the systemic delivery and endothelial penetration properties of AAV-PHP.B vectors. We discuss how receptor affinity analysis can be used to isolate vectors with optimized properties, improve the interpretation of library selections, and ultimately translate vector activities between preclinical animal models and humans.
Collapse
Affiliation(s)
- R. Alexander Martino
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qiang Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hao Xu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gui Hu
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Edgardo J. Arroyo
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joshua J. Sims
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M. Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
4
|
Liu M, van Kuppeveld FJM, de Haan CAM, de Vries E. Gradual adaptation of animal influenza A viruses to human-type sialic acid receptors. Curr Opin Virol 2023; 60:101314. [DOI: 10.1016/j.coviro.2023.101314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
|
5
|
Hamming PHE, Overeem NJ, Diestelhorst K, Fiers T, Tieke M, Vos GM, Boons GJPH, van der Vries E, Block S, Huskens J. Receptor Density-Dependent Motility of Influenza Virus Particles on Surface Gradients. ACS APPLIED MATERIALS & INTERFACES 2023; 15:25066-25076. [PMID: 37167605 DOI: 10.1021/acsami.3c05299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Influenza viruses can move across the surface of host cells while interacting with their glycocalyx. This motility may assist in finding or forming locations for cell entry and thereby promote cellular uptake. Because the binding to and cleavage of cell surface receptors forms the driving force for the process, the surface-bound motility of influenza is expected to be dependent on the receptor density. Surface gradients with gradually varying receptor densities are thus a valuable tool to study binding and motility processes of influenza and can function as a mimic for local receptor density variations at the glycocalyx that may steer the directionality of a virus particle in finding the proper site of uptake. We have tracked individual influenza virus particles moving over surfaces with receptor density gradients. We analyzed the extracted virus tracks first at a general level to verify neuraminidase activity and subsequently with increasing detail to quantify the receptor density-dependent behavior on the level of individual virus particles. While a directional bias was not observed, most likely due to limitations of the steepness of the surface gradient, the surface mobility and the probability of sticking were found to be significantly dependent on receptor density. A combination of high surface mobility and high dissociation probability of influenza was observed at low receptor densities, while the opposite occurred at higher receptor densities. These properties result in an effective mechanism for finding high-receptor density patches, which are believed to be a key feature of potential locations for cell entry.
Collapse
Affiliation(s)
- P H Erik Hamming
- Molecular Nanofabrication Group, MESA+ Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Nico J Overeem
- Molecular Nanofabrication Group, MESA+ Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Kevin Diestelhorst
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Tren Fiers
- Molecular Nanofabrication Group, MESA+ Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Malte Tieke
- Division of Virology, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Gaël M Vos
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Geert-Jan P H Boons
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, 3584 CG Utrecht, The Netherlands
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Department of Chemistry, University of Georgia, Athens, Georgia 30602, United States
| | - Erhard van der Vries
- Division of Virology, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
- Royal GD, Arnsbergstraat 7, 7418 EZ Deventer, The Netherlands
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Stephan Block
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jurriaan Huskens
- Molecular Nanofabrication Group, MESA+ Institute, Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
6
|
Schirripa Spagnolo C, Luin S. Setting up multicolour TIRF microscopy down to the single molecule level. Biomol Concepts 2023; 14:bmc-2022-0032. [PMID: 37428621 DOI: 10.1515/bmc-2022-0032] [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: 05/11/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
Investigating biological mechanisms in ever greater detail requires continuous advances in microscopy techniques and setups. Total internal reflection fluorescence (TIRF) microscopy is a well-established technique for visualizing processes on the cell membrane. TIRF allows studies down to the single molecule level, mainly in single-colour applications. Instead, multicolour setups are still limited. Here, we describe our strategies for implementing a multi-channel TIRF microscopy system capable of simultaneous two-channel excitation and detection, starting from a single-colour commercial setup. First, we report some applications at high molecule density and then focus on the challenges we faced for achieving the single molecule level simultaneously in different channels, showing that rigorous optimizations on the setup are needed to increase its sensitivity up to this point, from camera setting to background minimization. We also discuss our strategies regarding crucial points of fluorescent labelling for this type of experiment: labelling strategy, kind of probe, efficiency, and orthogonality of the reaction, all of which are aspects that can influence the achievable results. This work may provide useful guidelines for setting up advanced single-molecule multi-channel TIRF experiments to obtain insights into interaction mechanisms on the cell membrane of living cells.
Collapse
Affiliation(s)
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, I-56127, Pisa, Italy
- NEST Laboratory, Istituto Nanoscienze-Consiglio Nazionale delle ricerche (CNR), Piazza San Silvestro 12, I-56127, Pisa, Italy
| |
Collapse
|
7
|
Casalino L, Seitz C, Lederhofer J, Tsybovsky Y, Wilson IA, Kanekiyo M, Amaro RE. Breathing and Tilting: Mesoscale Simulations Illuminate Influenza Glycoprotein Vulnerabilities. ACS CENTRAL SCIENCE 2022; 8:1646-1663. [PMID: 36589893 PMCID: PMC9801513 DOI: 10.1021/acscentsci.2c00981] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Indexed: 05/28/2023]
Abstract
Influenza virus has resurfaced recently from inactivity during the early stages of the COVID-19 pandemic, raising serious concerns about the nature and magnitude of future epidemics. The main antigenic targets of influenza virus are two surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA). Whereas the structural and dynamical properties of both glycoproteins have been studied previously, the understanding of their plasticity in the whole-virion context is fragmented. Here, we investigate the dynamics of influenza glycoproteins in a crowded protein environment through mesoscale all-atom molecular dynamics simulations of two evolutionary-linked glycosylated influenza A whole-virion models. Our simulations reveal and kinetically characterize three main molecular motions of influenza glycoproteins: NA head tilting, HA ectodomain tilting, and HA head breathing. The flexibility of HA and NA highlights antigenically relevant conformational states, as well as facilitates the characterization of a novel monoclonal antibody, derived from convalescent human donor, that binds to the underside of the NA head. Our work provides previously unappreciated views on the dynamics of HA and NA, advancing the understanding of their interplay and suggesting possible strategies for the design of future vaccines and antivirals against influenza.
Collapse
Affiliation(s)
- Lorenzo Casalino
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California92093, United States
| | - Christian Seitz
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California92093, United States
| | - Julia Lederhofer
- Vaccine
Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland20892, United States
| | - Yaroslav Tsybovsky
- Electron
Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research
Sponsored by the National Cancer Institute, Frederick, Maryland21702, United States
| | - Ian A. Wilson
- Department
of Integrative Structural and Computational Biology and the Skaggs
Institute for Chemical Biology, The Scripps
Research Institute, La Jolla, California92037, United States
| | - Masaru Kanekiyo
- Vaccine
Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland20892, United States
| | - Rommie E. Amaro
- Department
of Chemistry and Biochemistry, University
of California San Diego, La Jolla, California92093, United States
| |
Collapse
|
8
|
Armanious A, Gerelli Y, Micciulla S, Pace HP, Welbourn RJL, Sjöberg M, Agnarsson B, Höök F. Probing the Separation Distance between Biological Nanoparticles and Cell Membrane Mimics Using Neutron Reflectometry with Sub-Nanometer Accuracy. J Am Chem Soc 2022; 144:20726-20738. [DOI: 10.1021/jacs.2c08456] [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)
- Antonius Armanious
- Department of Physics, Chalmers University of Technology, 41296Gothenburg, Sweden
| | - Yuri Gerelli
- Institut Max von Laue-Paul Langevin (ILL), 38042Grenoble, France
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131Ancona, Italy
| | | | - Hudson P. Pace
- Department of Physics, Chalmers University of Technology, 41296Gothenburg, Sweden
| | - Rebecca J. L. Welbourn
- ISIS Facility, STFC, Rutherford Appleton Laboratory, Chilton, Didcot, OxonOX11 0QX, United Kingdom
| | - Mattias Sjöberg
- Department of Physics, Chalmers University of Technology, 41296Gothenburg, Sweden
| | - Björn Agnarsson
- Department of Physics, Chalmers University of Technology, 41296Gothenburg, Sweden
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, 41296Gothenburg, Sweden
| |
Collapse
|
9
|
Negi G, Sharma A, Dey M, Dhanawat G, Parveen N. Membrane attachment and fusion of HIV-1, influenza A, and SARS-CoV-2: resolving the mechanisms with biophysical methods. Biophys Rev 2022; 14:1109-1140. [PMID: 36249860 PMCID: PMC9552142 DOI: 10.1007/s12551-022-00999-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/16/2022] [Indexed: 10/31/2022] Open
Abstract
Attachment to and fusion with cell membranes are two major steps in the replication cycle of many human viruses. We focus on these steps for three enveloped viruses, i.e., HIV-1, IAVs, and SARS-CoV-2. Viral spike proteins drive the membrane attachment and fusion of these viruses. Dynamic interactions between the spike proteins and membrane receptors trigger their specific attachment to the plasma membrane of host cells. A single virion on cell membranes can engage in binding with multiple receptors of the same or different types. Such dynamic and multivalent binding of these viruses result in an optimal attachment strength which in turn leads to their cellular entry and membrane fusion. The latter process is driven by conformational changes of the spike proteins which are also class I fusion proteins, providing the energetics of membrane tethering, bending, and fusion. These viruses exploit cellular and membrane factors in regulating the conformation changes and membrane processes. Herein, we describe the major structural and functional features of spike proteins of the enveloped viruses including highlights on their structural dynamics. The review delves into some of the case studies in the literature discussing the findings on multivalent binding, membrane hemifusion, and fusion of these viruses. The focus is on applications of biophysical tools with an emphasis on single-particle methods for evaluating mechanisms of these processes at the molecular and cellular levels.
Collapse
Affiliation(s)
- Geetanjali Negi
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Anurag Sharma
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Manorama Dey
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Garvita Dhanawat
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| | - Nagma Parveen
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, India
| |
Collapse
|
10
|
Casalino L, Seitz C, Lederhofer J, Tsybovsky Y, Wilson IA, Kanekiyo M, Amaro RE. Breathing and tilting: mesoscale simulations illuminate influenza glycoprotein vulnerabilities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.02.502576. [PMID: 35982676 PMCID: PMC9387122 DOI: 10.1101/2022.08.02.502576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Influenza virus has resurfaced recently from inactivity during the early stages of the COVID-19 pandemic, raising serious concerns about the nature and magnitude of future epidemics. The main antigenic targets of influenza virus are two surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA). Whereas the structural and dynamical properties of both glycoproteins have been studied previously, the understanding of their plasticity in the whole-virion context is fragmented. Here, we investigate the dynamics of influenza glycoproteins in a crowded protein environment through mesoscale all-atom molecular dynamics simulations of two evolutionary-linked glycosylated influenza A whole-virion models. Our simulations reveal and kinetically characterize three main molecular motions of influenza glycoproteins: NA head tilting, HA ectodomain tilting, and HA head breathing. The flexibility of HA and NA highlights antigenically relevant conformational states, as well as facilitates the characterization of a novel monoclonal antibody, derived from human convalescent plasma, that binds to the underside of the NA head. Our work provides previously unappreciated views on the dynamics of HA and NA, advancing the understanding of their interplay and suggesting possible strategies for the design of future vaccines and antivirals against influenza.
Collapse
Affiliation(s)
- Lorenzo Casalino
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Christian Seitz
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Julia Lederhofer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, United States
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology and the Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, United States
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Rommie E. Amaro
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
11
|
Characterization of binding interactions of SARS-CoV-2 spike protein and DNA-peptide nanostructures. Sci Rep 2022; 12:12828. [PMID: 35896714 PMCID: PMC9328006 DOI: 10.1038/s41598-022-16914-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 07/15/2022] [Indexed: 12/22/2022] Open
Abstract
Binding interactions of the spike proteins of the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) to a peptide fragment derived from the human angiotensin converting enzyme 2 (hACE2) receptor are investigated. The peptide is employed as capture moiety in enzyme linked immunosorbent assays (ELISA) and quantitative binding interaction measurements that are based on fluorescence proximity sensing (switchSENSE). In both techniques, the peptide is presented on an oligovalent DNA nanostructure, in order to assess the impact of mono- versus trivalent binding modes. As the analyte, the spike protein and several of its subunits are tested as well as inactivated SARS-CoV-2 and pseudo viruses. While binding of the peptide to the full-length spike protein can be observed, the subunits RBD and S1 do not exhibit binding in the employed concentrations. Variations of the amino acid sequence of the recombinant full-length spike proteins furthermore influence binding behavior. The peptide was coupled to DNA nanostructures that form a geometric complement to the trimeric structure of the spike protein binding sites. An increase in binding strength for trimeric peptide presentation compared to single peptide presentation could be generally observed in ELISA and was quantified in switchSENSE measurements. Binding to inactivated wild type viruses could be shown as well as qualitatively different binding behavior of the Alpha and Beta variants compared to the wild type virus strain in pseudo virus models.
Collapse
|
12
|
Marbach S, Holmes-Cerfon M. Mass Changes the Diffusion Coefficient of Particles with Ligand-Receptor Contacts in the Overdamped Limit. PHYSICAL REVIEW LETTERS 2022; 129:048003. [PMID: 35939031 DOI: 10.1103/physrevlett.129.048003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/29/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
Inertia does not generally affect the long-time diffusion of passive overdamped particles in fluids. Yet a model starting from the Langevin equation predicts a surprising property of particles coated with ligands that bind reversibly to surface receptors: heavy particles diffuse more slowly than light ones of the same size. We show this by simulation and by deriving an analytic formula for the mass-dependent diffusion coefficient in the overdamped limit. We estimate the magnitude of this effect for a range of biophysical ligand-receptor systems, and find it is potentially observable for tailored micronscale DNA-coated colloids.
Collapse
Affiliation(s)
- Sophie Marbach
- Courant Institute of Mathematical Sciences, New York University, New York 10012, USA
- CNRS, Sorbonne Université, Physicochimie des Electrolytes et Nanosystèmes Interfaciaux, F-75005 Paris, France
| | - Miranda Holmes-Cerfon
- Courant Institute of Mathematical Sciences, New York University, New York 10012, USA
| |
Collapse
|
13
|
Liu M, Huang LZX, Smits AA, Büll C, Narimatsu Y, van Kuppeveld FJM, Clausen H, de Haan CAM, de Vries E. Human-type sialic acid receptors contribute to avian influenza A virus binding and entry by hetero-multivalent interactions. Nat Commun 2022; 13:4054. [PMID: 35831293 PMCID: PMC9279479 DOI: 10.1038/s41467-022-31840-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/06/2022] [Indexed: 11/09/2022] Open
Abstract
Establishment of zoonotic viruses, causing pandemics like the Spanish flu and Covid-19, requires adaptation to human receptors. Pandemic influenza A viruses (IAV) that crossed the avian-human species barrier switched from binding avian-type α2-3-linked sialic acid (2-3Sia) to human-type 2-6Sia receptors. Here, we show that this specificity switch is however less dichotomous as generally assumed. Binding and entry specificity were compared using mixed synthetic glycan gradients of 2-3Sia and 2-6Sia and by employing a genetically remodeled Sia repertoire on the surface of a Sia-free cell line and on a sialoglycoprotein secreted from these cells. Expression of a range of (mixed) 2-3Sia and 2-6Sia densities shows that non-binding human-type receptors efficiently enhanced avian IAV binding and entry provided the presence of a low density of high affinity avian-type receptors, and vice versa. Considering the heterogeneity of sialoglycan receptors encountered in vivo, hetero-multivalent binding is physiologically relevant and will impact evolutionary pathways leading to host adaptation. It is believed that human Influenza HA glycoprotein attaches to alpha2-6 linked sialic acids (SA) on cells, while avian viruses bind to alpha2-3 linked sialic acids, therewith contributing to host tropism. Here, Liu et al. show that mixing low-affinity alpha2-3 SA with low amounts of high-affinity alpha2-6 SA increases binding and entry of human viruses and the converse for avian virus. This shows that receptor recognition is not as strict as currently assumed and provides evidence that heteromultivalent interactions between human/avian HA and SA contributes to host adaptation.
Collapse
Affiliation(s)
- Mengying Liu
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Liane Z X Huang
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Anthony A Smits
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Christian Büll
- Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark.,Department of Biomolecular Chemistry, Institute for Molecules and Materials, Radboud University, Nijmegen, The Netherlands
| | - Yoshiki Narimatsu
- Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Frank J M van Kuppeveld
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Henrik Clausen
- Center for Glycomics, Departments of Cellular and Molecular Medicine and Odontology, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, Copenhagen, Denmark
| | - Cornelis A M de Haan
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Erik de Vries
- Virology group, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Ruiz PAS, Ziebert F, Kulić IM. Physics of self-rolling viruses. Phys Rev E 2022; 105:054411. [PMID: 35706307 DOI: 10.1103/physreve.105.054411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/29/2022] [Indexed: 06/15/2023]
Abstract
Viruses are right at the interface of inanimate matter and life. However, recent experiments [Sakai et al., J. Virol. 92, e01522-17 (2018)0022-538X10.1128/JVI.01522-17] have shown that some influenza strains can actively roll on glycan-covered surfaces. In a previous letter [Ziebert and Kulić, Phys. Rev. Lett. 126, 218101 (2021)0031-900710.1103/PhysRevLett.126.218101] we suggested this to be a form of viral surface metabolism: a collection of spike proteins that attach to and cut the glycans act as a self-organized mechano-chemical motor. Here we study in more depth the physics of the emergent self-rolling states. We give scaling arguments how the motion arises, substantiated by a detailed analytical theory that yields the full torque-angular velocity relation of the self-organized motor. Stochastic Gillespie simulations are used to validate the theory and to quantify stochastic effects like virus detachment and reversals of its direction. Finally, we also cross-check several approximations made previously and show that the proposed mechanism is very robust. All these results point together to the statistical inevitability of viral rolling in the presence of enzymatic activity.
Collapse
Affiliation(s)
- Pedro A Soria Ruiz
- Institute for Theoretical Physics, Heidelberg University, D-69120 Heidelberg, Germany
| | - Falko Ziebert
- Institute for Theoretical Physics, Heidelberg University, D-69120 Heidelberg, Germany
- BioQuant, Heidelberg University, D-69120 Heidelberg, Germany
| | - Igor M Kulić
- Institut Charles Sadron UPR22-CNRS, F-67034 Strasbourg, France
- Institute Theory of Polymers, Leibniz-Institute of Polymer Research, D-01069 Dresden, Germany
| |
Collapse
|
15
|
Cui F, Marbach S, Zheng JA, Holmes-Cerfon M, Pine DJ. Comprehensive view of microscopic interactions between DNA-coated colloids. Nat Commun 2022; 13:2304. [PMID: 35484104 PMCID: PMC9051097 DOI: 10.1038/s41467-022-29853-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
The self-assembly of DNA-coated colloids into highly-ordered structures offers great promise for advanced optical materials. However, control of disorder, defects, melting, and crystal growth is hindered by the lack of a microscopic understanding of DNA-mediated colloidal interactions. Here we use total internal reflection microscopy to measure in situ the interaction potential between DNA-coated colloids with nanometer resolution and the macroscopic melting behavior. The range and strength of the interaction are measured and linked to key material design parameters, including DNA sequence, polymer length, grafting density, and complementary fraction. We present a first-principles model that screens and combines existing theories into one coherent framework and quantitatively reproduces our experimental data without fitting parameters over a wide range of DNA ligand designs. Our theory identifies a subtle competition between DNA binding and steric repulsion and accurately predicts adhesion and melting at a molecular level. Combining experimental and theoretical results, our work provides a quantitative and predictive approach for guiding material design with DNA-nanotechnology and can be further extended to a diversity of colloidal and biological systems. A quantitative prediction of DNA-mediated interactions between colloids is crucial to the design of colloidal structures for optical applications. Cui et al. measure the interaction potential with nanometer resolution and propose a theory to accurately predict adhesion and melting at a molecular level.
Collapse
Affiliation(s)
- Fan Cui
- Department of Physics, New York University, New York, NY, USA
| | - Sophie Marbach
- Courant Institute of Mathematical Sciences, New York University, New York, NY, USA.,CNRS, Sorbonne Université, Physicochimie des Electrolytes et Nanosystèmes, Interfaciaux, F-75005, Paris, France
| | | | | | - David J Pine
- Department of Physics, New York University, New York, NY, USA. .,Department of Chemical & Biomolecular Engineering, New York University, New York, NY, USA.
| |
Collapse
|
16
|
Marbach S, Zheng JA, Holmes-Cerfon M. The nanocaterpillar's random walk: diffusion with ligand-receptor contacts. SOFT MATTER 2022; 18:3130-3146. [PMID: 35348560 DOI: 10.1039/d1sm01544c] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Particles with ligand-receptor contacts bind and unbind fluctuating "legs" to surfaces, whose fluctuations cause the particle to diffuse. Quantifying the diffusion of such "nanoscale caterpillars" is a challenge, since binding events often occur on very short time and length scales. Here we derive an analytical formula, validated by simulations, for the long time translational diffusion coefficient of an overdamped nanocaterpillar, under a range of modeling assumptions. We demonstrate that the effective diffusion coefficient, which depends on the microscopic parameters governing the legs, can be orders of magnitude smaller than the background diffusion coefficient. Furthermore it varies rapidly with temperature, and reproduces the striking variations seen in existing data and our own measurements of the diffusion of DNA-coated colloids. Our model gives insight into the mechanism of motion, and allows us to ask: when does a nanocaterpillar prefer to move by sliding, where one leg is always linked to the surface, and when does it prefer to move by hopping, which requires all legs to unbind simultaneously? We compare a range of systems (viruses, molecular motors, white blood cells, protein cargos in the nuclear pore complex, bacteria such as Escherichia coli, and DNA-coated colloids) and present guidelines to control the mode of motion for materials design.
Collapse
Affiliation(s)
- Sophie Marbach
- Courant Institute of Mathematical Sciences, New York University, NY, 10012, USA.
- CNRS, Sorbonne Université, Physicochimie des Electrolytes et Nanosystèmes Interfaciaux, F-75005 Paris, France
| | | | | |
Collapse
|
17
|
Wang H, Zang Y, Zhao Y, Hao D, Kang Y, Zhang J, Zhang Z, Zhang L, Yang Z, Zhang S. Sequence Matching between Hemagglutinin and Neuraminidase through Sequence Analysis Using Machine Learning. Viruses 2022; 14:v14030469. [PMID: 35336876 PMCID: PMC8950662 DOI: 10.3390/v14030469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 01/27/2023] Open
Abstract
To date, many experiments have revealed that the functional balance between hemagglutinin (HA) and neuraminidase (NA) plays a crucial role in viral mobility, production, and transmission. However, whether and how HA and NA maintain balance at the sequence level needs further investigation. Here, we applied principal component analysis and hierarchical clustering analysis on thousands of HA and NA sequences of A/H1N1 and A/H3N2. We discovered significant coevolution between HA and NA at the sequence level, which is closely related to the type of host species and virus epidemic years. Furthermore, we propose a sequence-to-sequence transformer model (S2STM), which mainly consists of an encoder and a decoder that adopts a multi-head attention mechanism for establishing the mapping relationship between HA and NA sequences. The training results reveal that the S2STM can effectively realize the “translation” from HA to NA or vice versa, thereby building a relationship network between them. Our work combines unsupervised and supervised machine learning methods to identify the sequence matching between HA and NA, which will advance our understanding of IAVs’ evolution and also provide a novel idea for sequence analysis methods.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Zhiwei Yang
- Correspondence: (Z.Y.); (S.Z.); Tel.: +86-029-8266-8634 (Z.Y.); +86-029-8266-0915 (S.Z.)
| | - Shengli Zhang
- Correspondence: (Z.Y.); (S.Z.); Tel.: +86-029-8266-8634 (Z.Y.); +86-029-8266-0915 (S.Z.)
| |
Collapse
|
18
|
Park H, Sut TN, Yoon BK, Zhdanov VP, Kim JW, Cho NJ, Jackman JA. Multivalency-Induced Shape Deformation of Nanoscale Lipid Vesicles: Size-Dependent Membrane Bending Effects. J Phys Chem Lett 2022; 13:1480-1488. [PMID: 35129365 DOI: 10.1021/acs.jpclett.2c00090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The size of membrane-enveloped virus particles, exosomes, and lipid vesicles strongly impacts functional properties in biological and applied contexts. Multivalent ligand-receptor interactions involving nanoparticle shape deformation are critical to such functions, yet the corresponding effect of nanoparticle size remains largely elusive. Herein, using an indirect nanoplasmonic sensing approach, we investigated how the nanoscale size properties of ligand-modified lipid vesicles affect real-time binding interactions, especially vesicle deformation processes, with a receptor-modified, cell membrane-mimicking platform. Together with theoretical analyses, our findings reveal a pronounced, size-dependent transition in the membrane bending properties of nanoscale lipid vesicles between 60 and 180 nm in diameter. For smaller vesicles, a large membrane bending energy enhanced vesicle stiffness while the osmotic pressure energy was the dominant modulating factor for larger, less stiff vesicles. These findings advance our fundamental understanding of how nanoparticle size affects multivalency-induced nanoparticle shape deformation and can provide guidance for the design of biomimetic nanoparticles with tailored nanomechanical properties.
Collapse
Affiliation(s)
- Hyeonjin Park
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive, 637553 Singapore
| | | | - Bo Kyeong Yoon
- School of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, Republic of Korea
| | - Vladimir P Zhdanov
- Boreskov Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | | | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive, 637553 Singapore
| | | |
Collapse
|
19
|
Olsén E, Jõemetsa S, González A, Joyce P, Zhdanov VP, Midtvedt D, Höök F. Diffusion of Lipid Nanovesicles Bound to a Lipid Membrane Is Associated with the Partial-Slip Boundary Condition. NANO LETTERS 2021; 21:8503-8509. [PMID: 34403260 PMCID: PMC8517973 DOI: 10.1021/acs.nanolett.1c02092] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During diffusion of nanoparticles bound to a cellular membrane by ligand-receptor pairs, the distance to the laterally mobile interface is sufficiently short for their motion to depend not only on the membrane-mediated diffusivity of the tethers but also in a not yet fully understood manner on nanoparticle size and interfacial hydrodynamics. By quantifying diffusivity, velocity, and size of individual membrane-bound liposomes subjected to a hydrodynamic shear flow, we have successfully separated the diffusivity contributions from particle size and number of tethers. The obtained diffusion-size relations for synthetic and extracellular lipid vesicles are not well-described by the conventional no-slip boundary condition, suggesting partial slip as well as a significant diffusivity dependence on the distance to the lipid bilayer. These insights, extending the understanding of diffusion of biological nanoparticles at lipid bilayers, are of relevance for processes such as cellular uptake of viruses and lipid nanoparticles or labeling of cell-membrane-residing molecules.
Collapse
Affiliation(s)
- Erik Olsén
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Silver Jõemetsa
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Adrián González
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| | - Paul Joyce
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
- UniSA:
Clinical and Health Sciences, University
of South Australia, 5000 Adelaide, Australia
| | - Vladimir P. Zhdanov
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
- Boreskov
Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Daniel Midtvedt
- Department
of Physics, University of Gothenburg, SE-41296 Göteborg, Sweden
| | - Fredrik Höök
- Department
of Physics, Chalmers University of Technology, SE-41296 Göteborg, Sweden
| |
Collapse
|
20
|
Stadtmueller MN, Bhatia S, Kiran P, Hilsch M, Reiter-Scherer V, Adam L, Parshad B, Budt M, Klenk S, Sellrie K, Lauster D, Seeberger PH, Hackenberger CPR, Herrmann A, Haag R, Wolff T. Evaluation of Multivalent Sialylated Polyglycerols for Resistance Induction in and Broad Antiviral Activity against Influenza A Viruses. J Med Chem 2021; 64:12774-12789. [PMID: 34432457 DOI: 10.1021/acs.jmedchem.1c00794] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of multivalent sialic acid-based inhibitors active against a variety of influenza A virus (IAV) strains has been hampered by high genetic and structural variability of the targeted viral hemagglutinin (HA). Here, we addressed this challenge by employing sialylated polyglycerols (PGs). Efficacy of prototypic PGs was restricted to a narrow spectrum of IAV strains. To understand this restriction, we selected IAV mutants resistant to a prototypic multivalent sialylated PG by serial passaging. Resistance mutations mapped to the receptor binding site of HA, which was accompanied by altered receptor binding profiles of mutant viruses as detected by glycan array analysis. Specifying the inhibitor functionalization to 2,6-α-sialyllactose (SL) and adjusting the linker yielded a rationally designed inhibitor covering an extended spectrum of inhibited IAV strains. These results highlight the importance of integrating virological data with chemical synthesis and structural data for the development of sialylated PGs toward broad anti-influenza compounds.
Collapse
Affiliation(s)
- Marlena N Stadtmueller
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
| | - Sumati Bhatia
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Pallavi Kiran
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Malte Hilsch
- Institut für Biologie, Molekulare Biophysik, IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Valentin Reiter-Scherer
- Institut für Biologie, Molekulare Biophysik, IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Lutz Adam
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle Strasse 10, 13125 Berlin, Germany.,Institut für Chemie, Humboldt Universität zu Berlin, Brook-Taylor Str. 2, 12489 Berlin, Germany
| | - Badri Parshad
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Matthias Budt
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
| | - Simon Klenk
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle Strasse 10, 13125 Berlin, Germany.,Institut für Chemie, Humboldt Universität zu Berlin, Brook-Taylor Str. 2, 12489 Berlin, Germany
| | - Katrin Sellrie
- Department for Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Daniel Lauster
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.,Institut für Biologie, Molekulare Biophysik, IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Peter H Seeberger
- Department for Biomolecular Systems, Max Planck Institute for Colloids and Interfaces, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Christian P R Hackenberger
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Roessle Strasse 10, 13125 Berlin, Germany
| | - Andreas Herrmann
- Institut für Biologie, Molekulare Biophysik, IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestraße 10, 13353 Berlin, Germany
| |
Collapse
|
21
|
Bally M, Block S, Höök F, Larson G, Parveen N, Rydell GE. Physicochemical tools for studying virus interactions with targeted cell membranes in a molecular and spatiotemporally resolved context. Anal Bioanal Chem 2021; 413:7157-7178. [PMID: 34490501 PMCID: PMC8421089 DOI: 10.1007/s00216-021-03510-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Abstract
The objective of this critical review is to provide an overview of how emerging bioanalytical techniques are expanding our understanding of the complex physicochemical nature of virus interactions with host cell surfaces. Herein, selected model viruses representing both non-enveloped (simian virus 40 and human norovirus) and enveloped (influenza A virus, human herpes simplex virus, and human immunodeficiency virus type 1) viruses are highlighted. The technologies covered utilize a wide range of cell membrane mimics, from supported lipid bilayers (SLBs) containing a single purified host membrane component to SLBs derived from the plasma membrane of a target cell, which can be compared with live-cell experiments to better understand the role of individual interaction pairs in virus attachment and entry. These platforms are used to quantify binding strengths, residence times, diffusion characteristics, and binding kinetics down to the single virus particle and single receptor, and even to provide assessments of multivalent interactions. The technologies covered herein are surface plasmon resonance (SPR), quartz crystal microbalance with dissipation (QCM-D), dynamic force spectroscopy (DFS), total internal reflection fluorescence (TIRF) microscopy combined with equilibrium fluctuation analysis (EFA) and single particle tracking (SPT), and finally confocal microscopy using multi-labeling techniques to visualize entry of individual virus particles in live cells. Considering the growing scientific and societal needs for untangling, and interfering with, the complex mechanisms of virus binding and entry, we hope that this review will stimulate the community to implement these emerging tools and strategies in conjunction with more traditional methods. The gained knowledge will not only contribute to a better understanding of the virus biology, but may also facilitate the design of effective inhibitors to block virus entry.
Collapse
Affiliation(s)
- Marta Bally
- Department of Clinical Microbiology & Wallenberg Centre for Molecular Medicine, Umeå University, 901 85, Umeå, Sweden
| | - Stephan Block
- Department of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden.
| | - Göran Larson
- Department of Laboratory Medicine, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Bruna Stråket 16, 413 45, Gothenburg, Sweden.
| | - Nagma Parveen
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, 208016, India
| | - Gustaf E Rydell
- Department of Infectious Diseases, Sahlgrenska Academy at the University of Gothenburg, 413 46, Gothenburg, Sweden
| |
Collapse
|
22
|
Guo R, Li M, Zuo X. DNA Framework-Mediated Geometric Renormalization of Gold Nanoparticles on a Two-Dimensional Fluidic Membrane Interface. Chempluschem 2021; 86:1472-1475. [PMID: 34520133 DOI: 10.1002/cplu.202100344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/02/2021] [Indexed: 01/03/2023]
Abstract
The precise arrangement of single entity is a crucial objective of nanoscience and holds great promise in various fields such as biology and material science. In this work, we develop a "DNA framework-mediated geometric renormalization" (DFMGR) strategy to reassemble gold nanoparticles into specific geometric shapes on a 2-dimensional (2D) fluidic membrane interface. Cholesterol-modified AuNPs are randomly anchored on the supported lipid bilayer (SLB) via the cholesterol-lipid interaction. We demonstrate that AuNPs are laterally mobile on SLB and could be further rearranged into a specific geometric shape by DNA framework containing algebraically topological DNA arms. Using scanning electron microscope (SEM) imaging approach, simple geometric shapes, such as points assembled by monomers, line segments assembled by dimers, triangles assembled by trimers are visually presented. Interestingly, we found that the statistic angle (58.77°) and side length (12.21 nm) of triangles obtained from SEM images were both agreed well with the theoretical angle of 60° and side length of 12.58 nm. And the relative error of the angle calculated was as low as 0.33 %. These results indicated that the DFMGR strategy showed precise regulation ability for the AuNPs renormalization. We believe that DNA framework-mediated geometric renormalization strategy would be a powerful means for regulating ligand-receptor interactions in biosystems and for nanoparticle assembling in material science.
Collapse
Affiliation(s)
- Ruiyan Guo
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Min Li
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Xiaolei Zuo
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University
| |
Collapse
|
23
|
Park H, Sut TN, Yoon BK, Zhdanov VP, Cho NJ, Jackman JA. Unraveling How Multivalency Triggers Shape Deformation of Sub-100 nm Lipid Vesicles. J Phys Chem Lett 2021; 12:6722-6729. [PMID: 34263601 DOI: 10.1021/acs.jpclett.1c01510] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Multivalent ligand-receptor interactions are critical to the function of membrane-enveloped biological and biomimetic nanoparticles, yet resulting nanoparticle shape changes are rarely investigated. Using the localized surface plasmon resonance (LSPR) sensing technique, we tracked the attachment of biotinylated, sub-100 nm lipid vesicles to a streptavidin-functionalized supported lipid bilayer (SLB) and developed an analytical model to extract quantitative details about the vesicle-SLB contact region. The experimental results were supported by theoretical analyses of biotin-streptavidin complex formation and corresponding structural and energetic aspects of vesicle deformation. Our findings reveal how varying the surface densities of streptavidin receptors in the SLB and biotin ligands in the vesicles affects the extent of nanometer-scale vesicle deformation. We also identify conditions, i.e., a critical ligand density, at which appreciable vesicle deformation began, which provides insight into how the membrane bending energy partially counterposes the multivalent binding interaction energy. These findings are generalizable to various multivalent ligand-receptor systems.
Collapse
Affiliation(s)
- Hyeonjin Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive 637553, Singapore
| | - Tun Naw Sut
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive 637553, Singapore
| | - Bo Kyeong Yoon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Vladimir P Zhdanov
- Boreskov Institute of Catalysis, Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nam-Joon Cho
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Drive 637553, Singapore
| | - Joshua A Jackman
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
24
|
Abstract
We present here a model for multivalent diffusive transport whereby a central point-like hub is coupled to multiple feet, which bind to complementary sites on a two-dimensional landscape. The available number of binding interactions is dependent on the number of feet (multivalency) and on their allowed distance from the central hub (span). Using Monte Carlo simulations that implement the Gillespie algorithm, we simulate multivalent diffusive transport processes for 100 distinct walker designs. Informed by our simulation results, we derive an analytical expression for the diffusion coefficient of a general multivalent diffusive process as a function of multivalency, span, and dissociation constant Kd. Our findings can be used to guide the experimental design of multivalent transporters, in particular, providing insight into how to overcome trade-offs between diffusivity and processivity.
Collapse
Affiliation(s)
- Antonia Kowalewski
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Nancy R Forde
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| | - Chapin S Korosec
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
25
|
Overeem NJ, Hamming PH(E, Tieke M, van der Vries E, Huskens J. Multivalent Affinity Profiling: Direct Visualization of the Superselective Binding of Influenza Viruses. ACS NANO 2021; 15:8525-8536. [PMID: 33978406 PMCID: PMC8158855 DOI: 10.1021/acsnano.1c00166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/05/2021] [Indexed: 05/23/2023]
Abstract
The influenza A virus (IAV) interacts with the glycocalyx of host cells through its surface proteins hemagglutinin (HA) and neuraminidase (NA). Quantitative biophysical measurements of these interactions may help to understand these interactions at the molecular level with the long-term aim to predict influenza infectivity and answer other biological questions. We developed a method, called multivalent affinity profiling (MAP), to measure virus binding profiles on receptor density gradients to determine the threshold receptor density, which is a quantitative measure of virus avidity toward a receptor. Here, we show that imaging of IAVs on receptor density gradients allows the direct visualization and efficient assessment of their superselective binding. We show how the multivalent binding of IAVs can be quantitatively assessed using MAP if the receptor density gradients are prepared around the threshold receptor density without crowding at the higher densities. The threshold receptor density increases strongly with increasing flow rate, showing that the superselective binding of IAV is influenced by shear force. This method of visualization and quantitative assessment of superselective binding allows not only comparative studies of IAV-receptor interactions, but also more fundamental studies of how superselectivity arises and is influenced by experimental conditions.
Collapse
Affiliation(s)
- Nico J. Overeem
- Department
of Molecules & Materials, MESA+ Institute for Nanotechnology,
Faculty of Science and Technology, University
of Twente, 7500 AE Enschede, The Netherlands
| | - P. H. (Erik) Hamming
- Department
of Molecules & Materials, MESA+ Institute for Nanotechnology,
Faculty of Science and Technology, University
of Twente, 7500 AE Enschede, The Netherlands
| | - Malte Tieke
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Erhard van der Vries
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
- Royal
GD, Arnsbergstraat 7, 7418 EZ, Deventer, The Netherlands
- Department
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Jurriaan Huskens
- Department
of Molecules & Materials, MESA+ Institute for Nanotechnology,
Faculty of Science and Technology, University
of Twente, 7500 AE Enschede, The Netherlands
| |
Collapse
|
26
|
Overeem NJ, van der Vries E, Huskens J. A Dynamic, Supramolecular View on the Multivalent Interaction between Influenza Virus and Host Cell. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007214. [PMID: 33682339 DOI: 10.1002/smll.202007214] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/11/2021] [Indexed: 06/12/2023]
Abstract
Understanding how influenza viruses traverse the mucus and recognize host cells is critical for evaluating their zoonotic potential, and for prevention and treatment of the disease. The surface of the influenza A virus is covered with the receptor-binding protein hemagglutinin and the receptor-cleaving enzyme neuraminidase, which jointly control the interactions between the virus and the host cell. These proteins are organized in closely spaced trimers and tetramers to facilitate multivalent interactions with sialic acid-terminated glycans. This review shows that the individually weak multivalent interactions of influenza viruses allow superselective binding, virus-induced recruitment of receptors, and the formation of dynamic complexes that facilitate molecular walking. Techniques to measure the avidity and receptor specificity of influenza viruses are reviewed, and the pivotal role of multivalent interactions with their emergent properties in crossing the mucus and entering host cells is discussed. A model is proposed for the initiation of cell entry through virus-induced receptor clustering. The multivalent interactions of influenza viruses are maintained in a dynamic regime by a functional balance between binding and cleaving.
Collapse
Affiliation(s)
- Nico J Overeem
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede, 7500 AE, The Netherlands
| | - Erhard van der Vries
- Royal GD, Arnsbergstraat 7, Deventer, 7418 EZ, The Netherlands
- Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, Utrecht, 3584CX, The Netherlands
| | - Jurriaan Huskens
- Department of Molecules and Materials, MESA+ Institute for Nanotechnology, Faculty of Science and Technology, University of Twente, P.O. Box 217, Enschede, 7500 AE, The Netherlands
| |
Collapse
|
27
|
Trexler M, Brusatori M, Auner G. Avidin-biotin complex-based capture coating platform for universal Influenza virus immobilization and characterization. PLoS One 2021; 16:e0247429. [PMID: 33635877 PMCID: PMC7909696 DOI: 10.1371/journal.pone.0247429] [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: 11/18/2020] [Accepted: 02/08/2021] [Indexed: 11/18/2022] Open
Abstract
Influenza virus mutates quickly and unpredictably creating emerging pathogenic strains that are difficult to detect, diagnose, and characterize. Conventional tools to study and characterize virus, such as next generation sequencing, genome amplification (RT-PCR), and serological antibody testing, are not adequately suited to rapidly mutating pathogens like Influenza virus where the success of infection heavily depends on the phenotypic expression of surface glycoproteins. Bridging the gap between genome and pathogenic expression remains a challenge. Using sialic acid as a universal Influenza virus binding receptor, a novel virus avidin-biotin complex-based capture coating was developed and characterized that may be used to create future diagnostic and interrogation platforms for viable whole Influenza virus. First, fluorescent FITC probe studies were used to optimize coating component concentrations. Then atomic force microscopy (AFM) was used to profile the surface characteristics of the novel capture coating, acquire topographical imaging of Influenza particles immobilized by the coating, and calculate the capture efficiency of the coating (over 90%) for all four representative human Influenza virus strains tested.
Collapse
Affiliation(s)
- Micaela Trexler
- Smart Sensors and Integrated Microsystems, Wayne State University, Detroit, Michigan, United States of America
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, Michigan, United States of America
- * E-mail:
| | - Michelle Brusatori
- Smart Sensors and Integrated Microsystems, Wayne State University, Detroit, Michigan, United States of America
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, Michigan, United States of America
- Michael and Marian Illitch Department of Surgery, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Gregory Auner
- Smart Sensors and Integrated Microsystems, Wayne State University, Detroit, Michigan, United States of America
- Department of Biomedical Engineering, Wayne State University College of Engineering, Detroit, Michigan, United States of America
- Michael and Marian Illitch Department of Surgery, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| |
Collapse
|
28
|
Korosec CS, Jindal L, Schneider M, Calderon de la Barca I, Zuckermann MJ, Forde NR, Emberly E. Substrate stiffness tunes the dynamics of polyvalent rolling motors. SOFT MATTER 2021; 17:1468-1479. [PMID: 33347523 DOI: 10.1039/d0sm01811b] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Nature has evolved many mechanisms for achieving directed motion on the subcellular level. The burnt-bridges ratchet (BBR) is one mechanism used to achieve superdiffusive molecular motion over long distances through the successive cleavage of surface-bound energy-rich substrate sites. This mechanism has been associated with both nanoscale and microscale movement, with the latter accomplished through polyvalent interactions between a large hub (e.g. influenza virus) and substrate (e.g. cell surface receptors). Experimental successes in achieving superdiffusive motion by synthetic polyvalent BBRs have raised questions about the dynamics of their motility, including whether rolling or translation is better able to direct motion of microscale spherical hubs. Here we simulate the three-dimensional dynamics of a polyvalent sphere moving on and cleaving an elastic substrate. We find that substrate stiffness plays an important role in controlling both the motor's mode of motility and its directional persistence. As we tune lateral substrate stiffness from soft to stiff we find there exists an intermediate value that optimizes rolling behaviour. We also find that there is an optimal substrate stiffness for maximizing persistence length, while stiffness does not influence as strongly the superdiffusive dynamics of the particle. Lastly, we examine the effect of substrate density, and show that softer landscapes are better able to buffer against decreases in substrate occupancy, with the spherical motor maintaining superdiffusive motion more on softer landscapes than on stiff landscapes as occupancy drops. Our results highlight the importance of surface in controlling the motion of polyvalent BBRs.
Collapse
Affiliation(s)
- Chapin S Korosec
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Lavisha Jindal
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Mathew Schneider
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Ignacio Calderon de la Barca
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Martin J Zuckermann
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Nancy R Forde
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| | - Eldon Emberly
- Department of Physics, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia V5A 1S6, Canada.
| |
Collapse
|
29
|
A quantitative view on multivalent nanomedicine targeting. Adv Drug Deliv Rev 2021; 169:1-21. [PMID: 33264593 DOI: 10.1016/j.addr.2020.11.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 11/11/2020] [Accepted: 11/21/2020] [Indexed: 12/17/2022]
Abstract
Although the concept of selective delivery has been postulated over 100 years ago, no targeted nanomedicine has been clinically approved so far. Nanoparticles modified with targeting ligands to promote the selective delivery of therapeutics towards a specific cell population have been extensively reported. However, the rational design of selective particles is still challenging. One of the main reasons for this is the lack of quantitative theoretical and experimental understanding of the interactions involved in cell targeting. In this review, we discuss new theoretical models and experimental methods that provide a quantitative view of targeting. We show the new advancements in multivalency theory enabling the rational design of super-selective nanoparticles. Furthermore, we present the innovative approaches to obtain key targeting parameters at the single-cell and single molecule level and their role in the design of targeting nanoparticles. We believe that the combination of new theoretical multivalent design and experimental methods to quantify receptors and ligands aids in the rational design and clinical translation of targeted nanomedicines.
Collapse
|
30
|
Nie C, Stadtmüller M, Parshad B, Wallert M, Ahmadi V, Kerkhoff Y, Bhatia S, Block S, Cheng C, Wolff T, Haag R. Heteromultivalent topology-matched nanostructures as potent and broad-spectrum influenza A virus inhibitors. SCIENCE ADVANCES 2021; 7:7/1/eabd3803. [PMID: 33523846 PMCID: PMC7775783 DOI: 10.1126/sciadv.abd3803] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 11/09/2020] [Indexed: 05/28/2023]
Abstract
Here, we report the topology-matched design of heteromultivalent nanostructures as potent and broad-spectrum virus entry inhibitors based on the host cell membrane. Initially, we investigate the virus binding dynamics to validate the better binding performance of the heteromultivalent moieties as compared to homomultivalent ones. The heteromultivalent binding moieties are transferred to nanostructures with a bowl-like shape matching the viral spherical surface. Unlike the conventional homomultivalent inhibitors, the heteromultivalent ones exhibit a half maximal inhibitory concentration of 32.4 ± 13.7 μg/ml due to the synergistic multivalent effects and the topology-matched shape. At a dose without causing cellular toxicity, >99.99% reduction of virus propagation has been achieved. Since multiple binding sites have also been identified on the S protein of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), we envision that the use of heteromultivalent nanostructures may also be applied to develop a potent inhibitor to prevent coronavirus infection.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Marlena Stadtmüller
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Badri Parshad
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, UK
| | - Matthias Wallert
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Vahid Ahmadi
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Yannic Kerkhoff
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Sumati Bhatia
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Stephan Block
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.
| | - Chong Cheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany.
| | - Rainer Haag
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany.
| |
Collapse
|
31
|
Anani T, Rahmati S, Sultana N, David AE. MRI-traceable theranostic nanoparticles for targeted cancer treatment. Am J Cancer Res 2021; 11:579-601. [PMID: 33391494 PMCID: PMC7738852 DOI: 10.7150/thno.48811] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Current cancer therapies, including chemotherapy and radiotherapy, are imprecise, non-specific, and are often administered at high dosages - resulting in side effects that severely impact the patient's overall well-being. A variety of multifunctional, cancer-targeted nanotheranostic systems that integrate therapy, imaging, and tumor targeting functionalities in a single platform have been developed to overcome the shortcomings of traditional drugs. Among the imaging modalities used, magnetic resonance imaging (MRI) provides high resolution imaging of structures deep within the body and, in combination with other imaging modalities, provides complementary diagnostic information for more accurate identification of tumor characteristics and precise guidance of anti-cancer therapy. This review article presents a comprehensive assessment of nanotheranostic systems that combine MRI-based imaging (T1 MRI, T2 MRI, and multimodal imaging) with therapy (chemo-, thermal-, gene- and combination therapy), connecting a range of topics including hybrid treatment options (e.g. combined chemo-gene therapy), unique MRI-based imaging (e.g. combined T1-T2 imaging, triple and quadruple multimodal imaging), novel targeting strategies (e.g. dual magnetic-active targeting and nanoparticles carrying multiple ligands), and tumor microenvironment-responsive drug release (e.g. redox and pH-responsive nanomaterials). With a special focus on systems that have been tested in vivo, this review is an essential summary of the most advanced developments in this rapidly evolving field.
Collapse
|
32
|
Overeem NJ, Hamming PHE, Grant OC, Di Iorio D, Tieke M, Bertolino MC, Li Z, Vos G, de Vries RP, Woods RJ, Tito NB, Boons GJPH, van der Vries E, Huskens J. Hierarchical Multivalent Effects Control Influenza Host Specificity. ACS CENTRAL SCIENCE 2020; 6:2311-2318. [PMID: 33376792 PMCID: PMC7760459 DOI: 10.1021/acscentsci.0c01175] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 05/15/2023]
Abstract
Understanding how emerging influenza viruses recognize host cells is critical in evaluating their zoonotic potential, pathogenicity, and transmissibility between humans. The surface of the influenza virus is covered with hemagglutinin (HA) proteins that can form multiple interactions with sialic acid-terminated glycans on the host cell surface. This multivalent binding affects the selectivity of the virus in ways that cannot be predicted from the individual receptor-ligand interactions alone. Here, we show that the intrinsic structural and energetic differences between the interactions of avian- or human-type receptors with influenza HA translate from individual site affinity and orientation through receptor length and density on the surface into virus avidity and specificity. We introduce a method to measure virus avidity using receptor density gradients. We found that influenza viruses attached stably to a surface at receptor densities that correspond to a minimum number of approximately 8 HA-glycan interactions, but more interactions were required if the receptors were short and human-type. Thus, the avidity and specificity of influenza viruses for a host cell depend not on the sialic acid linkage alone but on a combination of linkage and the length and density of receptors on the cell surface. Our findings suggest that threshold receptor densities play a key role in virus tropism, which is a predicting factor for both their virulence and zoonotic potential.
Collapse
Affiliation(s)
- Nico J. Overeem
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - P. H. Erik Hamming
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Oliver C. Grant
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
| | - Daniele Di Iorio
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Malte Tieke
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - M. Candelaria Bertolino
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Zeshi Li
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gaël Vos
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Robert P. de Vries
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Robert J. Woods
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
- E-mail:
| | - Nicholas B. Tito
- Electric
Ant Lab, Science Park
106, 1098 XG Amsterdam, The Netherlands
| | - Geert-Jan P. H. Boons
- Complex
Carbohydrate Research Center, University
of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United
States
- Department
of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical
Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
- Bijvoet Center
for Biomolecular Research, Utrecht University, 3584 CH Utrecht, The Netherlands
- Department
of Chemistry, University of Georgia, Athens, Georgia 30602, United States
- E-mail:
| | - Erhard van der Vries
- Division
of Virology, Department of Infectious Diseases and Immunology, Faculty
of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
- Royal
GD, Arnsbergstraat 7, 7418 EZ Deventer, The Netherlands
- Department
of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
- E-mail:
| | - Jurriaan Huskens
- Molecular
Nanofabrication Group, MESA+ Institute for Nanotechnology, Faculty
of Science and Technology, University of
Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- E-mail:
| |
Collapse
|
33
|
Debets VE, Janssen LMC, Šarić A. Characterising the diffusion of biological nanoparticles on fluid and cross-linked membranes. SOFT MATTER 2020; 16:10628-10639. [PMID: 33084724 DOI: 10.1039/d0sm00712a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Tracing the motion of macromolecules, viruses, and nanoparticles adsorbed onto cell membranes is currently the most direct way of probing the complex dynamic interactions behind vital biological processes, including cell signalling, trafficking, and viral infection. The resulting trajectories are usually consistent with some type of anomalous diffusion, but the molecular origins behind the observed anomalous behaviour are usually not obvious. Here we use coarse-grained molecular dynamics simulations to help identify the physical mechanisms that can give rise to experimentally observed trajectories of nanoscopic objects moving on biological membranes. We find that diffusion on membranes of high fluidities typically results in normal diffusion of the adsorbed nanoparticle, irrespective of the concentration of receptors, receptor clustering, or multivalent interactions between the particle and membrane receptors. Gel-like membranes on the other hand result in anomalous diffusion of the particle, which becomes more pronounced at higher receptor concentrations. This anomalous diffusion is characterised by local particle trapping in the regions of high receptor concentrations and fast hopping between such regions. The normal diffusion is recovered in the limit where the gel membrane is saturated with receptors. We conclude that hindered receptor diffusivity can be a common reason behind the observed anomalous diffusion of viruses, vesicles, and nanoparticles adsorbed on cell and model membranes. Our results enable direct comparison with experiments and offer a new route for interpreting motility experiments on cell membranes.
Collapse
Affiliation(s)
- V E Debets
- Department of Applied Physics, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | | |
Collapse
|
34
|
Wallert M, Nie C, Anilkumar P, Abbina S, Bhatia S, Ludwig K, Kizhakkedathu JN, Haag R, Block S. Mucin-Inspired, High Molecular Weight Virus Binding Inhibitors Show Biphasic Binding Behavior to Influenza A Viruses. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2004635. [PMID: 33135314 DOI: 10.1002/smll.202004635] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/28/2020] [Indexed: 06/11/2023]
Abstract
Multivalent binding inhibitors are a promising new class of antivirals that prevent virus infections by inhibiting virus binding to cell membranes. The design of these inhibitors is challenging as many properties, for example, inhibitor size and functionalization with virus attachment factors, strongly influence the inhibition efficiency. Here, virus binding inhibitors are synthesized, the size and functionalization of which are inspired by mucins, which are naturally occurring glycosylated proteins with high molecular weight (MDa range) and interact efficiently with various viruses. Hyperbranched polyglycerols (hPGs) with molecular weights ranging between 10 and 2600 kDa are synthesized, thereby hitting the size of mucins and allowing for determining the impact of inhibitor size on the inhibition efficiency. The hPGs are functionalized with sialic acids and sulfates, as suggested from the structure of mucins, and their inhibition efficiency is determined by probing the inhibition of influenza A virus (IAV) binding to membranes using various methods. The largest, mucin-sized inhibitor shows potent inhibition at pm concentrations, while the inhibition efficiency decreases with decreasing the molecular weight. Interestingly, the concentration-dependent IAV inhibition shows a biphasic behavior, which is attributed to differences in the binding affinity of the inhibitors to the two IAV envelope proteins, neuraminidase, and hemagglutinin.
Collapse
Affiliation(s)
- Matthias Wallert
- Institute of Chemistry and Biochemistry, Emmy-Noether Group "Bionanointerfaces", Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry, Macromolecular Chemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Parambath Anilkumar
- Centre for Blood Research, Life Sciences Institute, Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Srinivas Abbina
- Centre for Blood Research, Life Sciences Institute, Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Sumati Bhatia
- Institute of Chemistry and Biochemistry, Macromolecular Chemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Kai Ludwig
- Research Center for Electron Microscopy and Core Facility BioSupraMol, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 36a, Berlin, 14195, Germany
| | - Jayachandran N Kizhakkedathu
- Centre for Blood Research, Life Sciences Institute, Department of Pathology and Laboratory Medicine, University of British Columbia, 2350 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Macromolecular Chemistry, Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| | - Stephan Block
- Institute of Chemistry and Biochemistry, Emmy-Noether Group "Bionanointerfaces", Freie Universität Berlin, Takustr. 3, Berlin, 14195, Germany
| |
Collapse
|
35
|
Heida R, Bhide YC, Gasbarri M, Kocabiyik Ö, Stellacci F, Huckriede ALW, Hinrichs WLJ, Frijlink HW. Advances in the development of entry inhibitors for sialic-acid-targeting viruses. Drug Discov Today 2020; 26:122-137. [PMID: 33099021 PMCID: PMC7577316 DOI: 10.1016/j.drudis.2020.10.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/13/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022]
Abstract
Over the past decades, several antiviral drugs have been developed to treat a range of infections. Yet the number of treatable viral infections is still limited, and resistance to current drug regimens is an ever-growing problem. Therefore, additional strategies are needed to provide a rapid cure for infected individuals. An interesting target for antiviral drugs is the process of viral attachment and entry into the cell. Although most viruses use distinct host receptors for attachment to the target cell, some viruses share receptors, of which sialic acids are a common example. This review aims to give an update on entry inhibitors for a range of sialic-acid-targeting viruses and provides insight into the prospects for those with broad-spectrum potential.
Collapse
Affiliation(s)
- Rick Heida
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| | - Yoshita C Bhide
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands; Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Matteo Gasbarri
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Özgün Kocabiyik
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Francesco Stellacci
- Institute of Materials, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Anke L W Huckriede
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713AV Groningen, The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands.
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, 9713AV Groningen, The Netherlands
| |
Collapse
|
36
|
Hugentobler KG, Heinrich D, Berg J, Heberle J, Brzezinski P, Schlesinger R, Block S. Lipid Composition Affects the Efficiency in the Functional Reconstitution of the Cytochrome c Oxidase. Int J Mol Sci 2020; 21:ijms21196981. [PMID: 32977390 PMCID: PMC7583929 DOI: 10.3390/ijms21196981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 11/16/2022] Open
Abstract
The transmembrane protein cytochrome c oxidase (CcO) is the terminal oxidase in the respiratory chain of many aerobic organisms and catalyzes the reduction of dioxygen to water. This process maintains an electrochemical proton gradient across the membrane hosting the oxidase. CcO is a well-established model enzyme in bioenergetics to study the proton-coupled electron transfer reactions and protonation dynamics involved in these processes. Its catalytic mechanism is subject to ongoing intense research. Previous research, however, was mainly focused on the turnover of oxygen and electrons in CcO, while studies reporting proton turnover rates of CcO, that is the rate of proton uptake by the enzyme, are scarce. Here, we reconstitute CcO from R. sphaeroides into liposomes containing a pH sensitive dye and probe changes of the pH value inside single proteoliposomes using fluorescence microscopy. CcO proton turnover rates are quantified at the single-enzyme level. In addition, we recorded the distribution of the number of functionally reconstituted CcOs across the proteoliposome population. Studies are performed using proteoliposomes made of native lipid sources, such as a crude extract of soybean lipids and the polar lipid extract of E. coli, as well as purified lipid fractions, such as phosphatidylcholine extracted from soybean lipids. It is shown that these lipid compositions have only minor effects on the CcO proton turnover rate, but can have a strong impact on the reconstitution efficiency of functionally active CcOs. In particular, our experiments indicate that efficient functional reconstitution of CcO is strongly promoted by the addition of anionic lipids like phosphatidylglycerol and cardiolipin.
Collapse
Affiliation(s)
- Katharina Gloria Hugentobler
- Institute of Chemistry and Biochemistry, Emmy-Noether Group “Bionanointerfaces”, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany;
| | - Dorothea Heinrich
- Department of Physics, Genetic Biophysics, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany; (D.H.); (R.S.)
| | - Johan Berg
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden; (J.B.); (P.B.)
| | - Joachim Heberle
- Department of Physics, Experimental Molecular Biophysics, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany;
| | - Peter Brzezinski
- Department of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural Sciences, Stockholm University, SE-106 91 Stockholm, Sweden; (J.B.); (P.B.)
| | - Ramona Schlesinger
- Department of Physics, Genetic Biophysics, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany; (D.H.); (R.S.)
| | - Stephan Block
- Institute of Chemistry and Biochemistry, Emmy-Noether Group “Bionanointerfaces”, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany;
- Correspondence:
| |
Collapse
|
37
|
Nie C, Parshad B, Bhatia S, Cheng C, Stadtmüller M, Oehrl A, Kerkhoff Y, Wolff T, Haag R. Topology-Matching Design of an Influenza-Neutralizing Spiky Nanoparticle-Based Inhibitor with a Dual Mode of Action. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 132:15662-15666. [PMID: 32836497 PMCID: PMC7276915 DOI: 10.1002/ange.202004832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 12/12/2022]
Abstract
In this study, we demonstrate the concept of "topology-matching design" for virus inhibitors. With the current knowledge of influenza A virus (IAV), we designed a nanoparticle-based inhibitor (nano-inhibitor) that has a matched nanotopology to IAV virions and shows heteromultivalent inhibitory effects on hemagglutinin and neuraminidase. The synthesized nano-inhibitor can neutralize the viral particle extracellularly and block its attachment and entry to the host cells. The virus replication was significantly reduced by 6 orders of magnitude in the presence of the reverse designed nano-inhibitors. Even when used 24 hours after the infection, more than 99.999 % inhibition is still achieved, which indicates such a nano-inhibitor might be a potent antiviral for the treatment of influenza infection.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
- Unit 17Robert Koch InstitutSeestr. 1013353BerlinGermany
| | - Badri Parshad
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sumati Bhatia
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Chong Cheng
- College of Polymer Science and EngineeringSichuan UniversityNo.24 South Section 1, Yihuan Road610065ChengduChina
| | | | - Alexander Oehrl
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Yannic Kerkhoff
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | | | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
38
|
Nie C, Parshad B, Bhatia S, Cheng C, Stadtmüller M, Oehrl A, Kerkhoff Y, Wolff T, Haag R. Topology-Matching Design of an Influenza-Neutralizing Spiky Nanoparticle-Based Inhibitor with a Dual Mode of Action. Angew Chem Int Ed Engl 2020; 59:15532-15536. [PMID: 32421225 PMCID: PMC7497169 DOI: 10.1002/anie.202004832] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Indexed: 11/09/2022]
Abstract
In this study, we demonstrate the concept of "topology-matching design" for virus inhibitors. With the current knowledge of influenza A virus (IAV), we designed a nanoparticle-based inhibitor (nano-inhibitor) that has a matched nanotopology to IAV virions and shows heteromultivalent inhibitory effects on hemagglutinin and neuraminidase. The synthesized nano-inhibitor can neutralize the viral particle extracellularly and block its attachment and entry to the host cells. The virus replication was significantly reduced by 6 orders of magnitude in the presence of the reverse designed nano-inhibitors. Even when used 24 hours after the infection, more than 99.999 % inhibition is still achieved, which indicates such a nano-inhibitor might be a potent antiviral for the treatment of influenza infection.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
- Unit 17Robert Koch InstitutSeestr. 1013353BerlinGermany
| | - Badri Parshad
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sumati Bhatia
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Chong Cheng
- College of Polymer Science and EngineeringSichuan UniversityNo.24 South Section 1, Yihuan Road610065ChengduChina
| | | | - Alexander Oehrl
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | - Yannic Kerkhoff
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| | | | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustr. 314195BerlinGermany
| |
Collapse
|
39
|
Jõemetsa S, Joyce P, Lubart Q, Mapar M, Celauro E, Agnarsson B, Block S, Bally M, Esbjörner EK, Jeffries GDM, Höök F. Independent Size and Fluorescence Emission Determination of Individual Biological Nanoparticles Reveals that Lipophilic Dye Incorporation Does Not Scale with Particle Size. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9693-9700. [PMID: 32787069 DOI: 10.1021/acs.langmuir.0c00941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Advancements in nanoparticle characterization techniques are critical for improving the understanding of how biological nanoparticles (BNPs) contribute to different cellular processes, such as cellular communication, viral infection, as well as various drug-delivery applications. Since BNPs are intrinsically heterogeneous, there is a need for characterization methods that are capable of providing information about multiple parameters simultaneously, preferably at the single-nanoparticle level. In this work, fluorescence microscopy was combined with surface-based two-dimensional flow nanometry, allowing for simultaneous and independent determination of size and fluorescence emission of individual BNPs. In this way, the dependence of the fluorescence emission of the commonly used self-inserting lipophilic dye 3,3'-dioctadecyl-5,5'-di(4-sulfophenyl)oxacarbocyanine (SP-DiO) could successfully be correlated with nanoparticle size for different types of BNPs, including synthetic lipid vesicles, lipid vesicles derived from cellular membrane extracts, and extracellular vesicles derived from human SH-SY5Y cell cultures; all vesicles had a radius, r, of ∼50 nm and similar size distributions. The results demonstrate that the dependence of fluorescence emission of SP-DiO on nanoparticle size varies significantly between the different types of BNPs, with the expected dependence on membrane area, r2, being observed for synthetic lipid vesicles, while a significant weaker dependence on size was observed for BNPs with more complex composition. The latter observation is attributed to a size-dependent difference in membrane composition, which may influence either the optical properties of the dye and/or the insertion efficiency, indicating that the fluorescence emission of this type of self-inserting dye may not be reliable for determining size or size distribution of BNPs with complex lipid compositions.
Collapse
Affiliation(s)
- Silver Jõemetsa
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
| | - Paul Joyce
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
| | - Quentin Lubart
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41296 Göteborg, Sweden
| | - Mokhtar Mapar
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
| | - Emanuele Celauro
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41296 Göteborg, Sweden
| | - Björn Agnarsson
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
| | - Stephan Block
- Department of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195 Berlin, Germany
| | - Marta Bally
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
- Department of Clinical Microbiology & Wallenberg Centre for Molecular Medicine, Umeå University, NUS Målpunkt R, 901 85 Umeå, Sweden
| | - Elin K Esbjörner
- Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, SE-41296 Göteborg, Sweden
| | - Gavin D M Jeffries
- Department of Chemistry and Chemical Engineering, Chalmers University of Technology, Kemivägen 10, SE-41296 Göteborg, Sweden
| | - Fredrik Höök
- Department of Physics, Chalmers University of Technology, Fysikgränd 3, SE-41296 Göteborg, Sweden
| |
Collapse
|
40
|
Bhatia S, Hilsch M, Cuellar‐Camacho JL, Ludwig K, Nie C, Parshad B, Wallert M, Block S, Lauster D, Böttcher C, Herrmann A, Haag R. Adaptive Flexible Sialylated Nanogels as Highly Potent Influenza A Virus Inhibitors. Angew Chem Int Ed Engl 2020; 59:12417-12422. [PMID: 32441859 PMCID: PMC7384064 DOI: 10.1002/anie.202006145] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 11/13/2022]
Abstract
Flexible multivalent 3D nanosystems that can deform and adapt onto the virus surface via specific ligand-receptor multivalent interactions can efficiently block virus adhesion onto the cell. We here report on the synthesis of a 250 nm sized flexible sialylated nanogel that adapts onto the influenza A virus (IAV) surface via multivalent binding of its sialic acid (SA) residues with hemagglutinin spike proteins on the virus surface. We could demonstrate that the high flexibility of sialylated nanogel improves IAV inhibition by 400 times as compared to a rigid sialylated nanogel in the hemagglutination inhibition assay. The flexible sialylated nanogel efficiently inhibits the influenza A/X31 (H3N2) infection with IC50 values in low picomolar concentrations and also blocks the virus entry into MDCK-II cells.
Collapse
Affiliation(s)
- Sumati Bhatia
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Malte Hilsch
- Institute of Biology & IRI Life SciencesHumboldt-Universität zu BerlinInvalidenstraße 4210115BerlinGermany
| | | | - Kai Ludwig
- Forschungszentrum für Elektronenmikroskopie, and Core Facility BioSupraMolInstitute of Chemistry and BiochemistryFreie Universität BerlinFabeckstr. 36a14195BerlinGermany
| | - Chuanxiong Nie
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Badri Parshad
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Matthias Wallert
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Stephan Block
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Daniel Lauster
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| | - Christoph Böttcher
- Forschungszentrum für Elektronenmikroskopie, and Core Facility BioSupraMolInstitute of Chemistry and BiochemistryFreie Universität BerlinFabeckstr. 36a14195BerlinGermany
| | - Andreas Herrmann
- Institute of Biology & IRI Life SciencesHumboldt-Universität zu BerlinInvalidenstraße 4210115BerlinGermany
| | - Rainer Haag
- Institute of Chemistry and BiochemistryFreie Universität BerlinTakustraße 314195BerlinGermany
| |
Collapse
|
41
|
Nie C, Stadtmüller M, Yang H, Xia Y, Wolff T, Cheng C, Haag R. Spiky Nanostructures with Geometry-matching Topography for Virus Inhibition. NANO LETTERS 2020; 20:5367-5375. [PMID: 32515974 DOI: 10.1021/acs.nanolett.0c01723] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Geometry-matching has been known to benefit the formation of stable biological interactions in natural systems. Herein, we report that the spiky nanostructures with matched topography to the influenza A virus (IAV) virions could be used to design next-generation advanced virus inhibitors. We demonstrated that nanostructures with spikes between 5 and 10 nm bind significantly better to virions than smooth nanoparticles, due to the short spikes inserting into the gaps of glycoproteins of the IAV virion. Furthermore, an erythrocyte membrane (EM) was coated to target the IAV, and the obtained EM-coated nanostructures could efficiently prevent IAV virion binding to the cells and inhibit subsequent infection. In a postinfection study, the EM-coated nanostructures reduced >99.9% virus replication at the cellular nontoxic dosage. We predict that such a combination of geometry-matching topography and cellular membrane coating will also push forward the development of nanoinhibitors for other virus strains, including SARS-CoV-2.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Marlena Stadtmüller
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Hua Yang
- Institute of Mechanics, Chair of Continuum Mechanics and Constitutive Theory, Technische Universität Berlin, Einsteinufer 5, 10587 Berlin, Germany
| | - Yi Xia
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| | - Thorsten Wolff
- Unit 17, Influenza and Other Respiratory Viruses, Robert Koch-Institut, Seestr. 10, 13353 Berlin, Germany
| | - Chong Cheng
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China
| | - Rainer Haag
- Institut für Chemie und Biochemie Organische Chemie, Freie Universität Berlin, Takustr. 3, 14195 Berlin, Germany
| |
Collapse
|
42
|
Bhatia S, Hilsch M, Cuellar‐Camacho JL, Ludwig K, Nie C, Parshad B, Wallert M, Block S, Lauster D, Böttcher C, Herrmann A, Haag R. Adaptive Flexible Sialylated Nanogels as Highly Potent Influenza A Virus Inhibitors. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Sumati Bhatia
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Malte Hilsch
- Institute of Biology & IRI Life Sciences Humboldt-Universität zu Berlin Invalidenstraße 42 10115 Berlin Germany
| | | | - Kai Ludwig
- Forschungszentrum für Elektronenmikroskopie, and Core Facility BioSupraMol Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 36a 14195 Berlin Germany
| | - Chuanxiong Nie
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Badri Parshad
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Matthias Wallert
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Stephan Block
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Daniel Lauster
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| | - Christoph Böttcher
- Forschungszentrum für Elektronenmikroskopie, and Core Facility BioSupraMol Institute of Chemistry and Biochemistry Freie Universität Berlin Fabeckstr. 36a 14195 Berlin Germany
| | - Andreas Herrmann
- Institute of Biology & IRI Life Sciences Humboldt-Universität zu Berlin Invalidenstraße 42 10115 Berlin Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry Freie Universität Berlin Takustraße 3 14195 Berlin Germany
| |
Collapse
|
43
|
Cuellar-Camacho JL, Bhatia S, Reiter-Scherer V, Lauster D, Liese S, Rabe JP, Herrmann A, Haag R. Quantification of Multivalent Interactions between Sialic Acid and Influenza A Virus Spike Proteins by Single-Molecule Force Spectroscopy. J Am Chem Soc 2020; 142:12181-12192. [PMID: 32538085 DOI: 10.1021/jacs.0c02852] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Multivalency is a key principle in reinforcing reversible molecular interactions through the formation of multiple bonds. The influenza A virus deploys this strategy to bind strongly to cell surface receptors. We performed single-molecule force spectroscopy (SMFS) to investigate the rupture force required to break individual and multiple bonds formed between synthetic sialic acid (SA) receptors and the two principal spike proteins of the influenza A virus (H3N2): hemagglutinin (H3) and neuraminidase (N2). Kinetic parameters such as the rupture length (χβ) and dissociation rate (koff) are extracted using the model by Friddle, De Yoreo, and Noy. We found that a monovalent SA receptor binds to N2 with a significantly higher bond lifetime (270 ms) compared to that for H3 (36 ms). By extending the single-bond rupture analysis to a multibond system of n protein-receptor pairs, we provide an unprecedented quantification of the mechanistic features of multivalency between H3 and N2 with SA receptors and show that the stability of the multivalent connection increases with the number of bonds from tens to hundreds of milliseconds. Association rates (kon) are also provided, and an estimation of the dissociation constants (KD) between the SA receptors to both proteins indicate a 17-fold higher binding affinity for the SA-N2 bond with respect to that of SA-H3. An optimal designed multivalent SA receptor showed a higher binding stability to the H3 protein of the influenza A virus than to the monovalent SA receptor. Our study emphasizes the influence of the scaffold on the presentation of receptors during multivalent binding.
Collapse
Affiliation(s)
- Jose Luis Cuellar-Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Sumati Bhatia
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| | - Valentin Reiter-Scherer
- Department of Physics & IRIS Adlershof, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Daniel Lauster
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany.,Institute for Biology & IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany
| | - Susanne Liese
- Department of Mathematics, University of Oslo, Moltke Moes vei 35, 1053 Oslo, Norway
| | - Jürgen P Rabe
- Department of Physics & IRIS Adlershof, Humboldt-Universität zu Berlin, Newtonstraße 15, 12489 Berlin, Germany
| | - Andreas Herrmann
- Institute for Biology & IRI Life Sciences, Humboldt-Universität zu Berlin, Invalidenstraße 42, 10115 Berlin, Germany
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195 Berlin, Germany
| |
Collapse
|
44
|
Delaveris CS, Webster ER, Banik SM, Boxer SG, Bertozzi CR. Membrane-tethered mucin-like polypeptides sterically inhibit binding and slow fusion kinetics of influenza A virus. Proc Natl Acad Sci U S A 2020; 117:12643-12650. [PMID: 32457151 PMCID: PMC7293601 DOI: 10.1073/pnas.1921962117] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The mechanism(s) by which cell-tethered mucins modulate infection by influenza A viruses (IAVs) remain an open question. Mucins form both a protective barrier that can block virus binding and recruit IAVs to bind cells via the sialic acids of cell-tethered mucins. To elucidate the molecular role of mucins in flu pathogenesis, we constructed a synthetic glycocalyx to investigate membrane-tethered mucins in the context of IAV binding and fusion. We designed and synthesized lipid-tethered glycopolypeptide mimics of mucins and added them to lipid bilayers, allowing chemical control of length, glycosylation, and surface density of a model glycocalyx. We observed that the mucin mimics undergo a conformational change at high surface densities from a compact to an extended architecture. At high surface densities, asialo mucin mimics inhibited IAV binding to underlying glycolipid receptors, and this density correlated to the mucin mimic's conformational transition. Using a single virus fusion assay, we observed that while fusion of virions bound to vesicles coated with sialylated mucin mimics was possible, the kinetics of fusion was slowed in a mucin density-dependent manner. These data provide a molecular model for a protective mechanism by mucins in IAV infection, and therefore this synthetic glycocalyx provides a useful reductionist model for studying the complex interface of host-pathogen interactions.
Collapse
Affiliation(s)
| | | | - Steven M Banik
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Steven G Boxer
- Department of Chemistry, Stanford University, Stanford, CA 94305;
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305;
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| |
Collapse
|
45
|
Steinmetz NF, Lim S, Sainsbury F. Protein cages and virus-like particles: from fundamental insight to biomimetic therapeutics. Biomater Sci 2020; 8:2771-2777. [PMID: 32352101 PMCID: PMC8085892 DOI: 10.1039/d0bm00159g] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Protein cages (viral and non-viral) found in nature have evolved for a variety of purposes and are found in all kingdoms of life. The main functions of these nanoscale compartments are the protection and delivery of nucleic acids e.g. virus capsids, or the enrichment and sequestration of metabolons e.g. bacterial microcompartments. This review focuses on recent developments of protein cages for use in immunotherapy and therapeutic delivery. In doing so, we highlight the unique ways in which protein cages have informed on fundamental principles governing bio-nano interactions. With the enormous existing design space among naturally occurring protein cages, there is still much to learn from studying them as biomimetic particles.
Collapse
Affiliation(s)
- Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, CA 92093, USA and Department of Bioengineering, University of California, San Diego, CA 92093, USA and Department of Radiology, University of California, San Diego, CA 92093, USA and Moores Cancer Center, University of California, San Diego, CA 92093, USA and Center for Nano-ImmunoEngineering, University of California, San Diego, CA 92093, USA
| | - Sierin Lim
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637457, Singapore and NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore 637457, Singapore
| | - Frank Sainsbury
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia. and Synthetic Biology Future Science Platform, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Brisbane, QLD 4001, Australia
| |
Collapse
|
46
|
Curk T, Tito NB. First-order 'hyper-selective' binding transition of multivalent particles under force. JOURNAL OF PHYSICS. CONDENSED MATTER : AN INSTITUTE OF PHYSICS JOURNAL 2020; 32:214002. [PMID: 31952055 DOI: 10.1088/1361-648x/ab6d12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Multivalent particles bind to targets via many independent ligand-receptor bonding interactions. This microscopic design spans length scales in both synthetic and biological systems. Classic examples include interactions between cells, virus binding, synthetic ligand-coated micrometer-scale vesicles or smaller nano-particles, functionalised polymers, and toxins. Equilibrium multivalent binding is a continuous yet super-selective transition with respect to the number of ligands and receptors involved in the interaction. Increasing the ligand or receptor density on the two particles leads to sharp growth in the number of bound particles at equilibrium. Here we present a theory and Monte Carlo simulations to show that applying mechanical force to multivalent particles causes their adsorption/desorption isotherm on a surface to become sharper and more selective, with respect to variation in the number of ligands and receptors on the two objects. When the force is only applied to particles bound to the surface by one or more ligands, then the transition can become infinitely sharp and first-order-a new binding regime which we term 'hyper-selective'. Force may be imposed by, e.g. flow of solvent around the particles, a magnetic field, chemical gradients, or triggered uncoiling of inert oligomers/polymers tethered to the particles to provide a steric repulsion to the surface. This physical principle is a step towards 'all or nothing' binding selectivity in the design of multivalent constructs.
Collapse
Affiliation(s)
- Tine Curk
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL 60208, United States of America
| | | |
Collapse
|
47
|
Koehler M, Delguste M, Sieben C, Gillet L, Alsteens D. Initial Step of Virus Entry: Virion Binding to Cell-Surface Glycans. Annu Rev Virol 2020; 7:143-165. [PMID: 32396772 DOI: 10.1146/annurev-virology-122019-070025] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Virus infection is an intricate process that requires the concerted action of both viral and host cell components. Entry of viruses into cells is initiated by interactions between viral proteins and cell-surface receptors. Various cell-surface glycans function as initial, usually low-affinity attachment factors, providing a first anchor of the virus to the cell surface, and further facilitate high-affinity binding to virus-specific cell-surface receptors, while other glycans function as specific entry receptors themselves. It is now possible to rapidly identify specific glycan receptors using different techniques, define atomic-level structures of virus-glycan complexes, and study these interactions at the single-virion level. This review provides a detailed overview of the role of glycans in viral infection and highlights experimental approaches to study virus-glycan binding along with specific examples. In particular, we highlight the development of the atomic force microscope to investigate interactions with glycans at the single-virion level directly on living mammalian cells, which offers new perspectives to better understand virus-glycan interactions in physiologically relevant conditions.
Collapse
Affiliation(s)
- Melanie Koehler
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium;
| | - Martin Delguste
- Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium;
| | - Christian Sieben
- Institute of Physics, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Laurent Gillet
- Immunology-Vaccinology Laboratory, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals and Health center (FARAH), University of Liège, 4000 Liège, 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
|
48
|
Dai X, Zhang X, Ostrikov K, Abrahamyan L. Host receptors: the key to establishing cells with broad viral tropism for vaccine production. Crit Rev Microbiol 2020; 46:147-168. [PMID: 32202955 PMCID: PMC7113910 DOI: 10.1080/1040841x.2020.1735992] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell culture-based vaccine technology is a flexible and convenient approach for vaccine production that requires adaptation of the vaccine strains to the new cells. Driven by the motivation to develop a broadly permissive cell line for infection with a wide range of viruses, we identified a set of the most relevant host receptors involved in viral attachment and entry. This identification was done through a review of different viral entry pathways and host cell lines, and in the context of the Baltimore classification of viruses. In addition, we indicated the potential technical problems and proposed some solutions regarding how to modify the host cell genome in order to meet industrial requirements for mass production of antiviral vaccines. Our work contributes to a finer understanding of the importance of breaking the host–virus recognition specificities for the possibility of creating a cell line feasible for the production of vaccines against a broad spectrum of viruses.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xuanhao Zhang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Kostya Ostrikov
- School of Chemistry and Physics and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Levon Abrahamyan
- Faculty of Veterinary Medicine, Swine and Poultry Infectious Diseases Research Center (CRIPA), Research Group on Infectious Diseases in Production Animals (GREMIP), Université de Montréal, Saint-Hyacinthe, Canada
| |
Collapse
|
49
|
Abstract
In recent decades, single particle tracking (SPT) has been developed into a sophisticated analytical approach involving complex instruments and data analysis schemes to extract information from time-resolved particle trajectories. Very often, mobility-related properties are extracted from these particle trajectories, as they often contain information about local interactions experienced by the particles while moving through the sample. This tutorial aims to provide a comprehensive overview about the accuracies that can be achieved when extracting mobility-related properties from 2D particle trajectories and how these accuracies depend on experimental parameters. Proper interpretation of SPT data requires an assessment of whether the obtained accuracies are sufficient to resolve the effect under investigation. This is demonstrated by calculating mean square displacement curves that show an apparent super- or subdiffusive behavior due to poor measurement statistics instead of the presence of true anomalous diffusion. Furthermore, the refinement of parameters involved in the design or analysis of SPT experiments is discussed and an approach is proposed in which square displacement distributions are inspected to evaluate the quality of SPT data and to extract information about the maximum distance over which particles should be tracked during the linking process.
Collapse
|
50
|
Hamming PHE, Overeem NJ, Huskens J. Influenza as a molecular walker. Chem Sci 2020; 11:27-36. [PMID: 32153750 PMCID: PMC7021193 DOI: 10.1039/c9sc05149j] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
The surface of the influenza virus is decorated with the receptor-binding protein hemagglutinin (HA) and the receptor-cleaving enzyme neuraminidase (NA). HA is responsible for host cell recognition, while NA prevents aggregation and entrapment, but the intricate mechanism of how the functions of these glycoproteins cooperate and how they are regulated by mutational responses to environmental pressures remains unclear. Recently, several groups have described the motion of influenza over surfaces and reported that this motion is inhibited by NA inhibitors. We argue that the motion of influenza resembles the motility of artificial receptor-cleaving particles called "molecular spiders". The cleaving of receptors by this type of molecular walkers leads to self-avoiding motion across a surface. When the binding and cleaving rates of molecular spiders are balanced, they move both rapidly and efficiently. The studies of molecular spiders offer new insights into the functional balance of HA and NA, but they do not address the asymmetric distribution of HA and NA on the surface of influenza. We propose that receptor-cleaving molecular walkers could play an important role in the further investigation of the motility of influenza viruses.
Collapse
Affiliation(s)
- P H Erik Hamming
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| | - Nico J Overeem
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| | - Jurriaan Huskens
- Molecular Nanofabrication Group , MESA + Institute for Nanotechnology , Faculty of Science and Technology , University of Twente , P.O. Box 217 , 7500 AE Enschede , The Netherlands .
| |
Collapse
|