1
|
Petroušková P, Bhide K, Mochnáčová E, Kulkarni A, Jozefiaková J, Tkáčová Z, Maľarik T, Kucková K, Talpašová L, Víglaský J, Kevély Á, Koči K, Nováková E, Koči J, Bhide M. Peptides developed against receptor binding sites of the E glycoprotein neutralize tick-borne encephalitis virus. Sci Rep 2025; 15:11435. [PMID: 40181101 PMCID: PMC11968884 DOI: 10.1038/s41598-025-95449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025] Open
Abstract
Infection caused by tick-borne encephalitis virus (TBEV) often manifests with meningitis or encephalitis. Domain III (DIII) of the envelope (E) glycoprotein on the TBEV surface facilitates virion attachment to the cell receptor and initiates cell entry. As a result, this research focused on the DIII to develop antiviral molecules that could prevent cell entry. We first identified two receptor-binding sites (RBS, 300SGLTYTMCDK309 and 317APTDSGHDTVVMEVTFSGTKPCR339) on DIII, that are involved in binding to human brain microvascular endothelial cells (hBMECs). Then we sought to isolate peptides from two combinatorial peptide-phage libraries that bind to the RBS and prevent protein E from attaching to hBMECs. Three cyclic peptides (CP2-CNSSKLHMC, CP20-CDGRPDRAC, CP23-CMKESIRGC) and a linear peptide (LP16-AFHPRQMETQMY) inhibited DIII binding to hBMECs. CP2 and CP20, both non-cytotoxic and hemocompatible, effectively neutralized a live virus. CP2 and CP20 deserve further attention for their potential application as anti-TBEV therapeutics that prevent viral cell entry.
Collapse
Affiliation(s)
- Patrícia Petroušková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Evelína Mochnáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Amod Kulkarni
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
- Institute of Neuroimmunology of Slovak Academy of Sciences, 84510, Bratislava, Slovakia
| | - Jana Jozefiaková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Zuzana Tkáčová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Tomáš Maľarik
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Katarína Kucková
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Lea Talpašová
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Jakub Víglaský
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia
| | - Ádám Kevély
- Department of Virus Ecology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 84215, Bratislava, Slovakia
| | - Kamila Koči
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 84215, Bratislava, Slovakia
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia
| | - Eva Nováková
- Department of Virus Ecology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University, Ilkovičova 6, 84215, Bratislava, Slovakia
| | - Juraj Koči
- Department of Virus Ecology, Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 84505, Bratislava, Slovakia.
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 84506, Bratislava, Slovakia.
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, The University of Veterinary Medicine and Pharmacy in Košice, Komenského 73, 04181, Košice, Slovakia.
- Institute of Neuroimmunology of Slovak Academy of Sciences, 84510, Bratislava, Slovakia.
| |
Collapse
|
2
|
Rahman SMR, Alzan HF, Laughery JM, Bastos RG, Ueti MW, Suarez CE. Structural and antigenic characterization of Babesia Bovis HAP2 domains. Sci Rep 2025; 15:7781. [PMID: 40044720 PMCID: PMC11882828 DOI: 10.1038/s41598-025-91359-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
The tick-borne apicomplexan parasite Babesia bovis causes bovine babesiosis which leads to enormous food and economic losses around the world. The existing resources to manage this disease are limited and have pitfalls, therefore, introduction of new strategies is urgently needed. B. bovis reproduces sexually in the midgut of its tick vector. HAP2, a well conserved ancient protein, plays a crucial role in the gamete fusion of this parasite and is a strong candidate for developing transmission-blocking vaccines. We previously demonstrated that immunization of cattle with full size B. bovis HAP2 blocks transmission of the parasite by Rhipicephalus microplus. Understanding the conserved structural features and antigenicity of HAP2 protein and its domains will facilitate developing effective methods to control pathogen transmission. In this study, we analyzed and compared AlphaFold2-predicted 3D structure of B. bovis HAP2 with the well-characterized crystal structures of HAP2 of Chlamydomonas reinhardtii and Arabidopsis thaliana. The comparisons and structural analysis resulted in the definition of three domains' sequences, fusion loops, and disulfide bonds in the B. bovis HAP2. In addition, recombinant versions of each three predicted HAP2 domains were recognized by antibodies from HAP2 immunized and transmission-protected cattle, confirming their antigenicity. Remarkably, domain II was highly recognized compared to the other two domains. This study introduces new directions in designing novel functional assays and improved vaccine design through targeting the HAP2 protein.
Collapse
Affiliation(s)
- S M Raihan Rahman
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Heba F Alzan
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- Parasitology and Animal Diseases Department, National Research Center, Dokki, Giza, Egypt
| | - Jacob M Laughery
- Animal Disease Research Unit, United States Department of Agriculture - Agricultural Research Service, Pullman, WA, USA
| | - Reginaldo G Bastos
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- Animal Disease Research Unit, United States Department of Agriculture - Agricultural Research Service, Pullman, WA, USA
| | - Massaro W Ueti
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
- Animal Disease Research Unit, United States Department of Agriculture - Agricultural Research Service, Pullman, WA, USA
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.
- Animal Disease Research Unit, United States Department of Agriculture - Agricultural Research Service, Pullman, WA, USA.
| |
Collapse
|
3
|
Yan K, Mao L, Lan J, Xiao Z. Advancements in dengue vaccines: A historical overview and pro-spects for following next-generation candidates. J Microbiol 2025; 63:e2410018. [PMID: 40044132 DOI: 10.71150/jm.2410018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/06/2025] [Indexed: 05/13/2025]
Abstract
Dengue, caused by four serotypes of dengue viruses (DENV-1 to DENV-4), is the most prevalent and widely mosquito-borne viral disease affecting humans. Dengue virus (DENV) infection has been reported in over 100 countries, and approximately half of the world's population is now at risk. The paucity of universally licensed DENV vaccines highlights the urgent need to address this public health concern. Action and atten-tion to antibody-dependent enhancement increase the difficulty of vaccine development. With the worsen-ing dengue fever epidemic, Dengvaxia® (CYD-TDV) and Qdenga® (TAK-003) have been approved for use in specific populations in affected areas. However, these vaccines do not provide a balanced immune response to all four DENV serotypes and the vaccination cannot cover all populations. There is still a need to develop a safe, broad-spectrum, and effective vaccine to address the increasing number of dengue cases worldwide. This review provides an overview of the existing DENV vaccines, as well as potential candidates for future studies on DENV vaccine development, and discusses the challenges and possible solutions in the field.
Collapse
Affiliation(s)
- Kai Yan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| | - Lingjing Mao
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection Chinese Academy of Sciences, Shanghai, P. R. China
- University of the Chinese Academy of Sciences, Beijing, P. R. China
| | - Jiaming Lan
- CAS Key Laboratory of Molecular Virology & Immunology, Shanghai Institute of Immunity and Infection Chinese Academy of Sciences, Shanghai, P. R. China
| | - Zhongdang Xiao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
4
|
Mackin SR, Sariol A, Diamond MS. Antibody-mediated control mechanisms of viral infections. Immunol Rev 2024; 328:205-220. [PMID: 39162394 PMCID: PMC11661935 DOI: 10.1111/imr.13383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Antibodies generated after vaccination or natural pathogen exposure are essential mediators of protection against many infections. Most studies with viruses have focused on antibody neutralization, in which protection is conferred by the fragment antigen binding region (Fab) through targeting of different steps in the viral lifecycle including attachment, internalization, fusion, and egress. Beyond neutralization, the fragment crystallizable (Fc) region of antibodies can integrate innate and adaptive immune responses by engaging complement components and distinct Fc gamma receptors (FcγR) on different host immune cells. In this review, we discuss recent advances in our understanding of antibody neutralization and Fc effector functions, and the assays used to measure them. Additionally, we describe the contexts in which these mechanisms are associated with protection against viruses and highlight how Fc-FcγR interactions can improve the potency of antibody-based therapies.
Collapse
Affiliation(s)
- Samantha R. Mackin
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
| | - Alan Sariol
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Pathology & Immunology and Center for Genome Sciences, Lab & Genomic Medicine, Washington University School of Medicine, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky the Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Castanha PMS, McEnaney PJ, Park Y, Bouwer A, Chaves EJF, Lins RD, Paciaroni NG, Dickson P, Carlson G, Cordeiro MT, Magalhaes T, Craigo J, Marques ETA, Kodadek T, Burke DS. Identification and characterization of a nonbiological small-molecular mimic of a Zika virus conformational neutralizing epitope. Proc Natl Acad Sci U S A 2024; 121:e2312755121. [PMID: 38743628 PMCID: PMC11127016 DOI: 10.1073/pnas.2312755121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Antigenic similarities between Zika virus (ZIKV) and other flaviviruses pose challenges to the development of virus-specific diagnostic tools and effective vaccines. Starting with a DNA-encoded one-bead-one-compound combinatorial library of 508,032 synthetic, non-natural oligomers, we selected and characterized small molecules that mimic ZIKV epitopes. High-throughput fluorescence-activated cell sorter-based bead screening was used to select molecules that bound IgG from ZIKV-immune but not from dengue-immune sera. Deep sequencing of the DNA from the "Zika-only" beads identified 40 candidate molecular structures. A lead candidate small molecule "CZV1-1" was selected that correctly identifies serum specimens from Zika-experienced patients with good sensitivity and specificity (85.3% and 98.4%, respectively). Binding competition studies of purified anti-CZV1-1 IgG against known ZIKV-specific monoclonal antibodies (mAbs) showed that CZV1-1 mimics a nonlinear, neutralizing conformational epitope in the domain III of the ZIKV envelope. Purified anti-CZV1-1 IgG neutralized infection of ZIKV in cell cultures with potencies comparable to highly specific ZIKV-neutralizing mAbs. This study demonstrates an innovative approach for identification of synthetic non-natural molecular mimics of conformational virus epitopes. Such molecular mimics may have value in the development of accurate diagnostic assays for Zika, as well as for other viruses.
Collapse
Affiliation(s)
- Priscila M. S. Castanha
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| | - Patrick J. McEnaney
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | - Yongseok Park
- Department of Biostatistics, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| | - Anthea Bouwer
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA15219
| | - Elton J. F. Chaves
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Roberto D. Lins
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | | | - Paige Dickson
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | | | - Marli T. Cordeiro
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Tereza Magalhaes
- Department of Entomology, Texas A&M University, College Station, TX77843
- Department of Preventive and Social Medicine, School of Medicine, Universidade Federal da Bahia, Bahia40026-010, Brazil
| | - Jodi Craigo
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, PA15219
| | - Ernesto T. A. Marques
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
- Department of Virology, Aggeu Magalhaes Institute, Oswaldo Cruz Foundation, Cidade Universitearia, Recife, Pernambuco50740-465, Brazil
| | - Thomas Kodadek
- Department of Chemistry, The Herbert Wertheim University of Florida Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL33458
| | - Donald S. Burke
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA15261
| |
Collapse
|
6
|
Banach M. Structural Outlier Detection and Zernike-Canterakis Moments for Molecular Surface Meshes-Fast Implementation in Python. Molecules 2023; 29:52. [PMID: 38202635 PMCID: PMC10779519 DOI: 10.3390/molecules29010052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Object retrieval systems measure the degree of similarity of the shape of 3D models. They search for the elements of the 3D model databases that resemble the query model. In structural bioinformatics, the query model is a protein tertiary/quaternary structure and the objective is to find similarly shaped molecules in the Protein Data Bank. With the ever-growing size of the PDB, a direct atomic coordinate comparison with all its members is impractical. To overcome this problem, the shape of the molecules can be encoded by fixed-length feature vectors. The distance of a protein to the entire PDB can be measured in this low-dimensional domain in linear time. The state-of-the-art approaches utilize Zernike-Canterakis moments for the shape encoding and supply the retrieval process with geometric data of the input structures. The BioZernike descriptors are a standard utility of the PDB since 2020. However, when trying to calculate the ZC moments locally, the issue of the deficiency of libraries readily available for use in custom programs (i.e., without relying on external binaries) is encountered, in particular programs written in Python. Here, a fast and well-documented Python implementation of the Pozo-Koehl algorithm is presented. In contrast to the more popular algorithm by Novotni and Klein, which is based on the voxelized volume, the PK algorithm produces ZC moments directly from the triangular surface meshes of 3D models. In particular, it can accept the molecular surfaces of proteins as its input. In the presented PK-Zernike library, owing to Numba's just-in-time compilation, a mesh with 50,000 facets is processed by a single thread in a second at the moment order 20. Since this is the first time the PK algorithm is used in structural bioinformatics, it is employed in a novel, simple, but efficient protein structure retrieval pipeline. The elimination of the outlying chain fragments via a fast PCA-based subroutine improves the discrimination ability, allowing for this pipeline to achieve an 0.961 area under the ROC curve in the BioZernike validation suite (0.997 for the assemblies). The correlation between the results of the proposed approach and of the 3D Surfer program attains values up to 0.99.
Collapse
Affiliation(s)
- Mateusz Banach
- Department of Bioinformatics and Telemedicine, Faculty of Medicine, Jagiellonian University Medical College, Medyczna 7, 30-688 Kraków, Poland
| |
Collapse
|
7
|
Sarker A, Dhama N, Gupta RD. Dengue virus neutralizing antibody: a review of targets, cross-reactivity, and antibody-dependent enhancement. Front Immunol 2023; 14:1200195. [PMID: 37334355 PMCID: PMC10272415 DOI: 10.3389/fimmu.2023.1200195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/20/2023] Open
Abstract
Dengue is the most common viral infection spread by mosquitoes, prevalent in tropical countries. The acute dengue virus (DENV) infection is a benign and primarily febrile illness. However, secondary infection with alternative serotypes can worsen the condition, leading to severe and potentially fatal dengue. The antibody raised by the vaccine or the primary infections are frequently cross-reactive; however, weakly neutralizing, and during subsequent infection, they may increase the odds of antibody-dependent enhancement (ADE). Despite that, many neutralizing antibodies have been identified against the DENV, which are thought to be useful in reducing dengue severity. Indeed, an antibody must be free from ADE for therapeutic application, as it is pretty common in dengue infection and escalates disease severity. Therefore, this review has described the critical characteristics of DENV and the potential immune targets in general. The primary emphasis is given to the envelope protein of DENV, where potential epitopes targeted for generating serotype-specific and cross-reactive antibodies have critically been described. In addition, a novel class of highly neutralizing antibodies targeted to the quaternary structure, similar to viral particles, has also been described. Lastly, we have discussed different aspects of the pathogenesis and ADE, which would provide significant insights into developing safe and effective antibody therapeutics and equivalent protein subunit vaccines.
Collapse
|
8
|
Stiasny K, Medits I, Roßbacher L, Heinz FX. Impact of structural dynamics on biological functions of flaviviruses. FEBS J 2023; 290:1973-1985. [PMID: 35246954 PMCID: PMC10952610 DOI: 10.1111/febs.16419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 11/30/2022]
Abstract
Flaviviruses comprise a number of mosquito- or tick-transmitted human pathogens of global public health importance. Advances in structural biology techniques have contributed substantially to our current understanding of the life cycle of these small enveloped RNA viruses and led to deep insights into details of virus assembly, maturation and cell entry. In addition to large-scale conformational changes and oligomeric rearrangements of envelope proteins during these processes, there is increasing evidence that smaller-scale protein dynamics (referred to as virus "breathing") can confer extra flexibility to these viruses for the fine-tuning of their interactions with the immune system and possibly with cellular factors they encounter in their complex ecological cycles in arthropod and vertebrate hosts. In this review, we discuss how work with tick-borne encephalitis virus has extended our view on flavivirus breathing, leading to the identification of a novel mechanism of antibody-mediated infection enhancement and demonstrating breathing intermediates of the envelope protein in the process of membrane fusion. These data are discussed in the context of other flaviviruses and the perspective of a potential role of virus breathing to cope with the requirements of adaptation and replication in evolutionarily very different hosts.
Collapse
Affiliation(s)
- Karin Stiasny
- Center for VirologyMedical University of ViennaAustria
| | - Iris Medits
- Center for VirologyMedical University of ViennaAustria
| | | | | |
Collapse
|
9
|
Dengue virus infection - a review of pathogenesis, vaccines, diagnosis and therapy. Virus Res 2023; 324:199018. [PMID: 36493993 DOI: 10.1016/j.virusres.2022.199018] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/19/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
The transmission of dengue virus (DENV) from an infected Aedes mosquito to a human, causes illness ranging from mild dengue fever to fatal dengue shock syndrome. The similar conserved structure and sequence among distinct DENV serotypes or different flaviviruses has resulted in the occurrence of cross reaction followed by antibody-dependent enhancement (ADE). Thus far, the vaccine which can provide effective protection against infection by different DENV serotypes remains the biggest hurdle to overcome. Therefore, deep investigation is crucial for the potent and effective therapeutic drugs development. In addition, the cross-reactivity of flaviviruses that leads to false diagnosis in clinical settings could result to delay proper intervention management. Thus, the accurate diagnostic with high specificity and sensitivity is highly required to provide prompt diagnosis in respect to render early treatment for DENV infected individuals. In this review, the recent development of neutralizing antibodies, antiviral agents, and vaccine candidates in therapeutic platform for DENV infection will be discussed. Moreover, the discovery of antigenic cryptic epitopes, principle of molecular mimicry, and application of single-chain or single-domain antibodies towards DENV will also be presented.
Collapse
|
10
|
Biner DW, Grosch JS, Ortoleva PJ. B-cell epitope discovery: The first protein flexibility-based algorithm-Zika virus conserved epitope demonstration. PLoS One 2023; 18:e0262321. [PMID: 36920995 PMCID: PMC10016673 DOI: 10.1371/journal.pone.0262321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 12/22/2021] [Indexed: 03/16/2023] Open
Abstract
Antibody-antigen interaction-at antigenic local environments called B-cell epitopes-is a prominent mechanism for neutralization of infection. Effective mimicry, and display, of B-cell epitopes is key to vaccine design. Here, a physical approach is evaluated for the discovery of epitopes which evolve slowly over closely related pathogens (conserved epitopes). The approach is 1) protein flexibility-based and 2) demonstrated with clinically relevant enveloped viruses, simulated via molecular dynamics. The approach is validated against 1) seven structurally characterized enveloped virus epitopes which evolved the least (out of thirty-nine enveloped virus-antibody structures), 2) two structurally characterized non-enveloped virus epitopes which evolved slowly (out of eight non-enveloped virus-antibody structures), and 3) eight preexisting epitope and peptide discovery algorithms. Rationale for a new benchmarking scheme is presented. A data-driven epitope clustering algorithm is introduced. The prediction of five Zika virus epitopes (for future exploration on recombinant vaccine technologies) is demonstrated. For the first time, protein flexibility is shown to outperform solvent accessible surface area as an epitope discovery metric.
Collapse
Affiliation(s)
- Daniel W. Biner
- Department of Chemistry, Indiana University, Bloomington, Indiana, United States of America
| | - Jason S. Grosch
- Department of Chemistry, Indiana University, Bloomington, Indiana, United States of America
| | - Peter J. Ortoleva
- Department of Chemistry, Indiana University, Bloomington, Indiana, United States of America
- * E-mail:
| |
Collapse
|
11
|
Marchi S, Dragoni F, Boccuto A, Idoko OT, Zazzi M, Sow S, Diallo A, Viviani S, Montomoli E, Vicenti I, Trombetta CM. Neutralizing activity of African lineage Zika virus immune sera towards Asian lineage. Acta Trop 2022; 237:106736. [DOI: 10.1016/j.actatropica.2022.106736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 11/27/2022]
|
12
|
In Silico Comparative Analysis of Predicted B Cell Epitopes against Dengue Virus (Serotypes 1–4) Isolated from the Philippines. Vaccines (Basel) 2022; 10:vaccines10081259. [PMID: 36016147 PMCID: PMC9415047 DOI: 10.3390/vaccines10081259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
Dengue is a viral mosquito-borne disease that rapidly spreads in tropical and subtropical countries, including the Philippines. One of its most distinguishing characteristics is the ability of the Dengue Virus (DENV) to easily surpass the innate responses of the body, thus activating B cells of the adaptive immunity to produce virus-specific antibodies. Moreover, Dengvaxia® is the only licensed vaccine for DENV, but recent studies showed that seronegative individuals become prone to increased disease severity and hospitalization. Owing to this limitation of the dengue vaccine, this study determined and compared consensus and unique B cell epitopes among each DENV (1–4) Philippine isolate to identify potential areas of interest for future vaccine studies and therapeutic developments. An in silico-based epitope prediction of forty (40) DENV 1–4 strains, each serotype represented by ten (10) sequences from The National Center for Biotechnology Information (NCBI), was conducted using Kolaskar and Tongaonkar antigenicity, Emini surface accessibility, and Parker hydrophilicity prediction in Immune Epitope Database (IEDB). Results showed that five (5) epitopes were consensus for DENV-1 with no detected unique epitope, one (1) consensus epitope for DENV-2 with two (2) unique epitopes, one (1) consensus epitope for DENV-3 plus two (2) unique epitopes, and two (2) consensus epitopes and one (1) unique epitope for DENV-4. The findings of this study would contribute to determining potential vaccine and diagnostic marker candidates for further research studies and immunological applications against DENV (1–4) Philippine isolates.
Collapse
|
13
|
Saipin K, Thaisomboonsuk B, Siridechadilok B, Chaitaveep N, Ramasoota P, Puttikhunt C, Sangiambut S, Jones A, Kraivong R, Sriburi R, Keelapang P, Sittisombut N, Junjhon J. A replication competent luciferase-secreting DENV2 reporter for sero-epidemiological surveillance of neutralizing and enhancing antibodies. J Virol Methods 2022; 308:114577. [PMID: 35843366 DOI: 10.1016/j.jviromet.2022.114577] [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: 03/27/2022] [Revised: 06/21/2022] [Accepted: 07/06/2022] [Indexed: 10/17/2022]
Abstract
Dengue virus (DENV) specific neutralizing and enhancing antibodies play crucial roles in dengue disease prevention and pathogenesis. DENV reporters are gaining popularity in the evaluation of these antibodies; their accessibility and acceptance may improve with more efficient production systems and indications of their antigenic equivalence to the wild-type virus. This study aimed to generate a replication competent luciferase-secreting DENV reporter (LucDENV2) and evaluate its feasibility in neutralizing and infection-enhancing antibody assays in comparison with wild-type DENV2, strain 16681, and a luciferase-secreting, single-round infectious DENV2 reporter (LucSIP). LucDENV2 replicated to similarly high levels as that of the parent 16681 virus in a commonly used mosquito cell line. LucDENV2 was neutralized in an antibody concentration-dependent manner by a monoclonal antibody specific to the flavivirus fusion loop and two antibodies specific to the E domain III, which closely resembled the neutralization patterns employing the LucSIP and wild-type DENV2. Parallel analysis of LucDENV2 and wild-type DENV2 revealed good agreement between the luciferase-based and focus-based neutralization and enhancement assays in a 96-well microplate format when employed against a set of clinical sera, suggesting comparable antigenic properties of LucDENV2 with those of the parent virus. The high-titer, replication competent, luciferase-secreting DENV reporter presented here should be a useful tool for fast and reliable quantitation of neutralizing and infection-enhancing antibodies in populations living in DENV-endemic areas.
Collapse
Affiliation(s)
- Krongkan Saipin
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand
| | - Butsaya Thaisomboonsuk
- Department of Virology, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok 10400, Thailand
| | - Bunpote Siridechadilok
- Frontier Biodesign and Bioengineering Research Team, National Center for Genetic Engineering and Biotechnology, Pathum-thani 12120, Thailand
| | - Nithinart Chaitaveep
- Royal Thai Army, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok 10400, Thailand
| | - Pongrama Ramasoota
- Center of Excellence for Antibody Research (CEAR), Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Chunya Puttikhunt
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum-thani 12120, Thailand; Division of Dengue Hemorrhagic Fever Research and Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sutha Sangiambut
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum-thani 12120, Thailand; Division of Dengue Hemorrhagic Fever Research and Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Anthony Jones
- Department of Virology, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok 10400, Thailand
| | - Romchat Kraivong
- Molecular Biology of Dengue and Flaviviruses Research Team, Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum-thani 12120, Thailand; Division of Dengue Hemorrhagic Fever Research and Siriraj Center of Research Excellence in Dengue and Emerging Pathogens, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Rungtawan Sriburi
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Poonsook Keelapang
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nopporn Sittisombut
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraphan Junjhon
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
14
|
Yan Y, Yang J, Xiao D, Yin J, Song M, Xu Y, Zhao L, Dai Q, Li Y, Wang C, Wang Z, Ren X, Yang X, Ni J, Liu M, Guo X, Li W, Chen X, Liu Z, Cao R, Zhong W. Nafamostat mesylate as a broad-spectrum candidate for the treatment of flavivirus infections by targeting envelope proteins. Antiviral Res 2022; 202:105325. [PMID: 35460703 DOI: 10.1016/j.antiviral.2022.105325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 01/24/2023]
Abstract
Epidemics caused by flaviviruses occur globally; however, no antiviral drugs treating flaviviruses infections have yet been developed. Nafamostat (NM) is a protease inhibitor approved for pancreatitis and anti-coagulation. The anti-flavivirus potential of NM has yet to be determined. Here, utilizing in vitro and in vivo infection assays, we present that NM effectively inhibits Zika virus (ZIKV) and other flaviviruses in vitro. NM inhibited the production of ZIKV viral RNA and proteins originating from Asia and African lineage in human-, mouse- and monkey-derived cell lines and the in vivo anti-ZIKV efficacy of NM was verified. Mode-of-action analysis using time-of-drug-addition assay, infectivity inhibition assay, surface plasmon resonance assay, and molecular docking revealed that NM interacted with viral particles and blocked the early stage of infection by targeting the domain III of ZIKV envelope protein. Analysing the anti-flavivirus effects of NM-related compounds suggested that the antiviral effect depended on the unique structure of NM. These findings suggest the potential use of NM as an anti-flavivirus candidate, and a novel drug design approach targeting the flavivirus envelope protein.
Collapse
Affiliation(s)
- Yunzheng Yan
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jingjing Yang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Dian Xiao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jiye Yin
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Mengwen Song
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Yijie Xu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Lei Zhao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Qingsong Dai
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yuexiang Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Cui Wang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Zhuang Wang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaofeng Ren
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xiaotong Yang
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Jie Ni
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Miaomiao Liu
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xiaojia Guo
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Wei Li
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Xingjuan Chen
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China; Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zhiqiang Liu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Ruiyuan Cao
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| | - Wu Zhong
- National Engineering Research Center for the Emergency Drug, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
| |
Collapse
|
15
|
Yu Y, Si L, Meng Y. Flavivirus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:171-197. [PMID: 35412141 DOI: 10.1007/978-981-16-8702-0_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Flaviviruses, including Dengue virus, Zika virus, Yellow fever virus, Japanese encephalitis virus, West Nile virus, cause thousands of deaths and millions of illnesses each year. The large outbreak of ZIKV in 2016 reminds us that flaviviruses can pose a serious threat to human safety and public health as emerging and re-emerging viruses. However, there are no specific drugs approved for the treatment of flavivirus infections. Due to no need to enter the cells, viral entry inhibitors have the unique advantage in suppressing viral infections. Flaviviruses bind to receptors and attach to the cell surface, then enter the endosome in a clathrin-dependent manner and finalizes the viral entry process after fusion with the cell membrane in a low pH environment. Small molecules, antibodies or peptides can inhibit flavivirus entry by targeting the above processes. Here, we focus on flavivirus entry inhibitors with well-defined target and antiviral activity. We hope that our review will provide a theoretical basis for flavivirus treatment and drug research and help to accelerate the clinical application of flavivirus entry inhibitors.
Collapse
Affiliation(s)
- Yufeng Yu
- Medical School, Nanjing University, Nanjing, Jiangsu, China.
| | - Lulu Si
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Meng
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Dali University, Dali, Yunnan, China
| |
Collapse
|
16
|
Anasir MI, Poh CL. Discovery of B-cell epitopes for development of dengue vaccines and antibody therapeutics. Med Microbiol Immunol 2022; 211:1-18. [PMID: 35059822 DOI: 10.1007/s00430-021-00726-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/04/2021] [Indexed: 10/19/2022]
Abstract
Dengue is one of the most frequently transmitted viral infections globally which creates a serious burden to the healthcare system in many countries in the tropical and subtropical regions. To date, no vaccine has demonstrated balanced protection against the four dengue serotypes. Dengvaxia as the only vaccine that has been licensed for use in endemic areas has shown an increased risk in dengue-naïve vaccines to develop severe dengue. A crucial element in protection from dengue infection is the neutralizing antibody responses. Therefore, the identification of protective linear B-cell epitopes can guide vaccine design and facilitate the development of monoclonal antibodies as dengue therapeutics. This review summarizes the identification of dengue B-cell epitopes within the envelope (E) protein of dengue that can be incorporated into peptide vaccine constructs. These epitopes have been identified through approaches such as bioinformatics, three-dimensional structure analysis of antibody-dengue complexes, mutagenesis/alanine scanning and escape mutant studies. Additionally, the therapeutic potential of monoclonal antibodies targeting the E protein of dengue is reviewed. This can provide a basis for the design of future dengue therapies.
Collapse
Affiliation(s)
- Mohd Ishtiaq Anasir
- Virology Unit, Infectious Disease Research Centre, Institute for Medical Research, National Institutes of Health, Setia Alam, Shah Alam, Selangor, Malaysia
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia
| | - Chit Laa Poh
- Centre for Virus and Vaccine Research, Sunway University, 5, Jalan Universiti, 47500, Bandar Sunway, Selangor, Malaysia.
| |
Collapse
|
17
|
Feng J, Dong X, DeCosta A, Su Y, Angrisano F, Sala KA, Blagborough AM, Lu C, Springer TA. Structural basis of malaria transmission blockade by a monoclonal antibody to gamete fusogen HAP2. eLife 2021; 10:74707. [PMID: 34939934 PMCID: PMC8806182 DOI: 10.7554/elife.74707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/03/2021] [Indexed: 11/13/2022] Open
Abstract
HAP2 is a transmembrane gamete fusogen found in multiple eukaryotic kingdoms and is structurally homologous to viral class II fusogens. Studies in Plasmodium have suggested that HAP2 is an attractive target for vaccines that block transmission of malaria. HAP2 has three extracellular domains, arranged in the order D2, D1, and D3. Here, we report monoclonal antibodies against the D3 fragment of Plasmodium berghei HAP2 and crystal structures of D3 in complex with Fab fragments of two of these antibodies, one of which blocks fertilization of Plasmodium berghei in vitro and transmission of malaria in mosquitoes. We also show how this Fab binds the complete HAP2 ectodomain with electron microscopy. The two antibodies cross-react with HAP2 among multiple plasmodial species. Our characterization of the Plasmodium D3 structure, HAP2 ectodomain architecture, and mechanism of inhibition provide insights for the development of a vaccine to block malaria transmission.
Collapse
Affiliation(s)
- Juan Feng
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | | | - Adam DeCosta
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | - Yang Su
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | | | | | | | - Chafen Lu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital
| | | |
Collapse
|
18
|
VanBlargan LA, Milutinovic PS, Goo L, DeMaso CR, Durbin AP, Whitehead SS, Pierson TC, Dowd KA. Dengue Virus Serotype 1 Conformational Dynamics Confers Virus Strain-Dependent Patterns of Neutralization by Polyclonal Sera. J Virol 2021; 95:e0095621. [PMID: 34549976 PMCID: PMC8577358 DOI: 10.1128/jvi.00956-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022] Open
Abstract
Dengue virus cocirculates globally as four serotypes (DENV1 to -4) that vary up to 40% at the amino acid level. Viral strains within a serotype further cluster into multiple genotypes. Eliciting a protective tetravalent neutralizing antibody response is a major goal of vaccine design, and efforts to characterize epitopes targeted by polyclonal mixtures of antibodies are ongoing. Previously, we identified two E protein residues (126 and 157) that defined the serotype-specific antibody response to DENV1 genotype 4 strain West Pac-74. DENV1 and DENV2 human vaccine sera neutralized DENV1 viruses incorporating these substitutions equivalently. In this study, we explored the contribution of these residues to the neutralization of DENV1 strains representing distinct genotypes. While neutralization of the genotype 1 strain TVP2130 was similarly impacted by mutation at E residues 126 and 157, mutation of these residues in the genotype 2 strain 16007 did not markedly change neutralization sensitivity, indicating the existence of additional DENV1 type-specific antibody targets. The accessibility of antibody epitopes can be strongly influenced by the conformational dynamics of virions and modified allosterically by amino acid variation. We found that changes at E domain II residue 204, shown previously to impact access to a poorly accessible E domain III epitope, impacted sensitivity of DENV1 16007 to neutralization by vaccine immune sera. Our data identify a role for minor sequence variation in changes to the antigenic structure that impacts antibody recognition by polyclonal immune sera. Understanding how the many structures sampled by flaviviruses influence antibody recognition will inform the design and evaluation of DENV immunogens. IMPORTANCE Dengue virus (DENV) is an important human pathogen that cocirculates globally as four serotypes. Because sequential infection by different DENV serotypes is associated with more severe disease, eliciting a protective neutralizing antibody response against all four serotypes is a major goal of vaccine efforts. Here, we report that neutralization of DENV serotype 1 by polyclonal antibody is impacted by minor sequence variation among virus strains. Our data suggest that mechanisms that control neutralization sensitivity extend beyond variation within antibody epitopes but also include the influence of single amino acids on the ensemble of structural states sampled by structurally dynamic virions. A more detailed understanding of the antibody targets of DENV-specific polyclonal sera and factors that govern their access to antibody has important implications for flavivirus antigen design and evaluation.
Collapse
Affiliation(s)
- Laura A. VanBlargan
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Biological Sciences Graduate Program, University of Maryland, College Park, Maryland, USA
| | - Pavle S. Milutinovic
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leslie Goo
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina R. DeMaso
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna P. Durbin
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Stephen S. Whitehead
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Theodore C. Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Kimberly A. Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
19
|
Pinto AK, Hassert M, Han X, Barker D, Carnelley T, Branche E, Steffen TL, Stone ET, Geerling E, Viramontes KM, Nykiforuk C, Toth D, Shresta S, Kodihalli S, Brien JD. The Ability of Zika virus Intravenous Immunoglobulin to Protect From or Enhance Zika Virus Disease. Front Immunol 2021; 12:717425. [PMID: 34552587 PMCID: PMC8450494 DOI: 10.3389/fimmu.2021.717425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022] Open
Abstract
The closely related flaviviruses, dengue and Zika, cause significant human disease throughout the world. While cross-reactive antibodies have been demonstrated to have the capacity to potentiate disease or mediate protection during flavivirus infection, the mechanisms responsible for this dichotomy are still poorly understood. To understand how the human polyclonal antibody response can protect against, and potentiate the disease in the context of dengue and Zika virus infection we used intravenous hyperimmunoglobulin (IVIG) preparations in a mouse model of the disease. Three IVIGs (ZIKV-IG, Control-Ig and Gamunex®) were evaluated for their ability to neutralize and/or enhance Zika, dengue 2 and 3 viruses in vitro. The balance between virus neutralization and enhancement provided by the in vitro neutralization data was used to predict the IVIG concentrations which could protect or enhance Zika, and dengue 2 disease in vivo. Using this approach, we were able to define the unique in vivo dynamics of complex polyclonal antibodies, allowing for both enhancement and protection from flavivirus infection. Our results provide a novel understanding of how polyclonal antibodies interact with viruses with implications for the use of polyclonal antibody therapeutics and the development and evaluation of the next generation flavivirus vaccines.
Collapse
Affiliation(s)
- Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Xiaobing Han
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | | | | | - Emilie Branche
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | - Tara L. Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Karla M. Viramontes
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - Derek Toth
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | - Sujan Shresta
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| |
Collapse
|
20
|
Huang Y, Williamson BD, Moodie Z, Carpp LN, Chambonneau L, DiazGranados CA, Gilbert PB. Analysis of Neutralizing Antibodies as a Correlate of Instantaneous Risk of Hospitalized Dengue in Placebo Recipients of Dengue Vaccine Efficacy Trials. J Infect Dis 2021; 225:332-340. [PMID: 34174082 DOI: 10.1093/infdis/jiab342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/24/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In the CYD14 (NCT01373281) and CYD15 (NCT01374516) dengue vaccine efficacy trials, Month 13 neutralizing antibody (nAb) titers correlated inversely with risk of symptomatic, virologically confirmed dengue (VCD) between Month 13 (one month post-final-dose) and Month 25. We assessed nAb titer as a correlate of instantaneous risk of hospitalized VCD (HVCD), for which participants were continually surveilled for 72 months. METHODS Using longitudinal nAb titers from the per-protocol immunogenicity subsets, we estimated hazard ratios (HRs) of HVCD by current nAb titer value for three correlate/endpoint pairs: average titer across all four serotypes/HVCD of any serotype (HVCD-Any), serotype-specific titer/homologous HVCD, and serotype-specific titer/heterologous HVCD. RESULTS Baseline-seropositive placebo recipients with higher average titer had lower instantaneous risk of HVCD-Any in 2-16-year-olds and in 9-16-year-olds (HR 0.26 or 0.15 per 10-fold increase in average titer by two methods, 95% CIs 0.14 to 0.45 and 0.07 to 0.34, respectively) pooled across both trials. Results were similar for homologous HVCD. There was evidence suggesting increased HVCD-Any risk in participants with low average titer (1:10 to 1:100) compared to seronegative participants (HR 1.85, 95% CI 0.93 to 3.68). CONCLUSIONS Natural infection-induced nAbs were inversely associated with hospitalized dengue, upon exceeding a relatively low threshold.
Collapse
Affiliation(s)
- Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America.,Department of Biostatistics, University of Washington, Seattle, 98109, United States of America
| | - Brian D Williamson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America
| | | | - Carlos A DiazGranados
- Clinical Sciences, Sanofi Pasteur, Swiftwater, Pennsylvania, United States of America
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, 98109, United States of America.,Department of Biostatistics, University of Washington, Seattle, 98109, United States of America
| |
Collapse
|
21
|
Fels JM, Maurer DP, Herbert AS, Wirchnianski AS, Vergnolle O, Cross RW, Abelson DM, Moyer CL, Mishra AK, Aguilan JT, Kuehne AI, Pauli NT, Bakken RR, Nyakatura EK, Hellert J, Quevedo G, Lobel L, Balinandi S, Lutwama JJ, Zeitlin L, Geisbert TW, Rey FA, Sidoli S, McLellan JS, Lai JR, Bornholdt ZA, Dye JM, Walker LM, Chandran K. Protective neutralizing antibodies from human survivors of Crimean-Congo hemorrhagic fever. Cell 2021; 184:3486-3501.e21. [PMID: 34077751 PMCID: PMC8559771 DOI: 10.1016/j.cell.2021.05.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/19/2021] [Accepted: 04/29/2021] [Indexed: 12/31/2022]
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a World Health Organization priority pathogen. CCHFV infections cause a highly lethal hemorrhagic fever for which specific treatments and vaccines are urgently needed. Here, we characterize the human immune response to natural CCHFV infection to identify potent neutralizing monoclonal antibodies (nAbs) targeting the viral glycoprotein. Competition experiments showed that these nAbs bind six distinct antigenic sites in the Gc subunit. These sites were further delineated through mutagenesis and mapped onto a prefusion model of Gc. Pairwise screening identified combinations of non-competing nAbs that afford synergistic neutralization. Further enhancements in neutralization breadth and potency were attained by physically linking variable domains of synergistic nAb pairs through bispecific antibody (bsAb) engineering. Although multiple nAbs protected mice from lethal CCHFV challenge in pre- or post-exposure prophylactic settings, only a single bsAb, DVD-121-801, afforded therapeutic protection. DVD-121-801 is a promising candidate suitable for clinical development as a CCHFV therapeutic.
Collapse
Affiliation(s)
- J Maximilian Fels
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Andrew S Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA; The Geneva Foundation, Tacoma, WA 98402, USA
| | - Ariel S Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Olivia Vergnolle
- Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Robert W Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | | | | | - Akaash K Mishra
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer T Aguilan
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ana I Kuehne
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | | | - Russell R Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Elisabeth K Nyakatura
- Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jan Hellert
- Structural Virology Unit, Department of Virology, CNRS UMR 3569, Institut Pasteur, Paris 75724, France
| | - Gregory Quevedo
- Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Leslie Lobel
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | | | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc., San Diego, CA 92121, USA
| | - Thomas W Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Felix A Rey
- Structural Virology Unit, Department of Virology, CNRS UMR 3569, Institut Pasteur, Paris 75724, France
| | - Simone Sidoli
- Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jonathan R Lai
- Deparment of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - John M Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA.
| | - Laura M Walker
- Adimab, LLC, Lebanon, NH 03766, USA; Adagio Therapeutics, Inc., Waltham, MA 02451, USA.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
22
|
VanBlargan LA, Errico JM, Kafai NM, Burgomaster KE, Jethva PN, Broeckel RM, Meade-White K, Nelson CA, Himansu S, Wang D, Handley SA, Gross ML, Best SM, Pierson TC, Fremont DH, Diamond MS. Broadly neutralizing monoclonal antibodies protect against multiple tick-borne flaviviruses. J Exp Med 2021; 218:e20210174. [PMID: 33831142 PMCID: PMC8040518 DOI: 10.1084/jem.20210174] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/30/2022] Open
Abstract
Although Powassan virus (POWV) is an emerging tick-transmitted flavivirus that causes severe or fatal neuroinvasive disease in humans, medical countermeasures have not yet been developed. Here, we developed a panel of neutralizing anti-POWV mAbs recognizing six distinct antigenic sites. The most potent of these mAbs bind sites within domain II or III of the envelope (E) protein and inhibit postattachment viral entry steps. A subset of these mAbs cross-react with other flaviviruses. Both POWV type-specific and cross-reactive neutralizing mAbs confer protection in mice against POWV infection when given as prophylaxis or postexposure therapy. Several cross-reactive mAbs mapping to either domain II or III also protect in vivo against heterologous tick-transmitted flaviviruses including Langat and tick-borne encephalitis virus. Our experiments define structural and functional correlates of antibody protection against POWV infection and identify epitopes targeted by broadly neutralizing antibodies with therapeutic potential against multiple tick-borne flaviviruses.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/immunology
- Cell Line
- Chlorocebus aethiops
- Cross Reactions/immunology
- Encephalitis Viruses, Tick-Borne/drug effects
- Encephalitis Viruses, Tick-Borne/genetics
- Encephalitis Viruses, Tick-Borne/immunology
- Encephalitis Viruses, Tick-Borne/physiology
- Encephalitis, Tick-Borne/immunology
- Encephalitis, Tick-Borne/prevention & control
- Encephalitis, Tick-Borne/virology
- Epitopes/immunology
- HEK293 Cells
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/immunology
- Mice, Inbred C57BL
- Mutation
- Vero Cells
- Viral Envelope Proteins/immunology
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
- Mice
Collapse
Affiliation(s)
- Laura A. VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John M. Errico
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Natasha M. Kafai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Katherine E. Burgomaster
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Rebecca M. Broeckel
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Kimberly Meade-White
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Christopher A. Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | | | - David Wang
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Scott A. Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | | | - Sonja M. Best
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Theodore C. Pierson
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Daved H. Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
23
|
Xie S, Zhang H, Liang Z, Yang X, Cao R. AXL, an Important Host Factor for DENV and ZIKV Replication. Front Cell Infect Microbiol 2021; 11:575346. [PMID: 33954117 PMCID: PMC8092360 DOI: 10.3389/fcimb.2021.575346] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Flaviviruses, as critically important pathogens, are still major public health problems all over the world. For instance, the evolution of ZIKV led to large-scale outbreaks in the Yap island in 2007. DENV was considered by the World Health Organization (WHO) as one of the 10 threats to global health in 2019. Enveloped viruses hijack a variety of host factors to complete its replication cycle. Phosphatidylserine (PS) receptor, AXL, is considered to be a candidate receptor for flavivirus invasion. In this review, we discuss the molecular structure of ZIKV and DENV, and how they interact with AXL to successfully invade host cells. A more comprehensive understanding of the molecular mechanisms of flavivirus-AXL interaction will provide crucial insights into the virus infection process and the development of anti-flavivirus therapeutics.
Collapse
Affiliation(s)
- Shengda Xie
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Huiru Zhang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhenjie Liang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xingmiao Yang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruibing Cao
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
24
|
Magar R, Yadav P, Barati Farimani A. Potential neutralizing antibodies discovered for novel corona virus using machine learning. Sci Rep 2021; 11:5261. [PMID: 33664393 PMCID: PMC7970853 DOI: 10.1038/s41598-021-84637-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/17/2021] [Indexed: 02/07/2023] Open
Abstract
The fast and untraceable virus mutations take lives of thousands of people before the immune system can produce the inhibitory antibody. The recent outbreak of COVID-19 infected and killed thousands of people in the world. Rapid methods in finding peptides or antibody sequences that can inhibit the viral epitopes of SARS-CoV-2 will save the life of thousands. To predict neutralizing antibodies for SARS-CoV-2 in a high-throughput manner, in this paper, we use different machine learning (ML) model to predict the possible inhibitory synthetic antibodies for SARS-CoV-2. We collected 1933 virus-antibody sequences and their clinical patient neutralization response and trained an ML model to predict the antibody response. Using graph featurization with variety of ML methods, like XGBoost, Random Forest, Multilayered Perceptron, Support Vector Machine and Logistic Regression, we screened thousands of hypothetical antibody sequences and found nine stable antibodies that potentially inhibit SARS-CoV-2. We combined bioinformatics, structural biology, and Molecular Dynamics (MD) simulations to verify the stability of the candidate antibodies that can inhibit SARS-CoV-2.
Collapse
Affiliation(s)
- Rishikesh Magar
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Prakarsh Yadav
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Amir Barati Farimani
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
25
|
Magar R, Yadav P, Barati Farimani A. Potential neutralizing antibodies discovered for novel corona virus using machine learning. Sci Rep 2021; 11:5261. [PMID: 33664393 DOI: 10.1101/2020.03.14.992156] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/17/2021] [Indexed: 05/22/2023] Open
Abstract
The fast and untraceable virus mutations take lives of thousands of people before the immune system can produce the inhibitory antibody. The recent outbreak of COVID-19 infected and killed thousands of people in the world. Rapid methods in finding peptides or antibody sequences that can inhibit the viral epitopes of SARS-CoV-2 will save the life of thousands. To predict neutralizing antibodies for SARS-CoV-2 in a high-throughput manner, in this paper, we use different machine learning (ML) model to predict the possible inhibitory synthetic antibodies for SARS-CoV-2. We collected 1933 virus-antibody sequences and their clinical patient neutralization response and trained an ML model to predict the antibody response. Using graph featurization with variety of ML methods, like XGBoost, Random Forest, Multilayered Perceptron, Support Vector Machine and Logistic Regression, we screened thousands of hypothetical antibody sequences and found nine stable antibodies that potentially inhibit SARS-CoV-2. We combined bioinformatics, structural biology, and Molecular Dynamics (MD) simulations to verify the stability of the candidate antibodies that can inhibit SARS-CoV-2.
Collapse
Affiliation(s)
- Rishikesh Magar
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Prakarsh Yadav
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA
| | - Amir Barati Farimani
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
- Machine Learning Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
26
|
Katzelnick LC, Bos S, Harris E. Protective and enhancing interactions among dengue viruses 1-4 and Zika virus. Curr Opin Virol 2020; 43:59-70. [PMID: 32979816 DOI: 10.1016/j.coviro.2020.08.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022]
Abstract
Dengue viruses 1-4 (DENV 1-4) and Zika virus (ZIKV) are closely related flaviviruses transmitted by Aedes mosquitoes that co-circulate in Asia, the Americas, Africa, and Oceania. Here, we review recent and historical literature on in vitro experiments, animal models, and clinical and epidemiological studies to describe how the sequence of DENV 1-4 and ZIKV infections modulates subsequent dengue and Zika disease outcome. Overall, we find these interactions are asymmetric. Immunity from a prior DENV infection or a prior ZIKV infection can enhance future severe dengue disease for some DENV serotypes while protecting against other serotypes. Further, prior DENV immunity has not been shown to enhance future uncomplicated or severe Zika and instead appears to be protective. Interestingly, secondary ZIKV infection induces type-specific ZIKV immunity but only generates weakly cross-neutralizing anti-DENV/ZIKV immunity, consistent with risk of future dengue disease. In contrast, secondary DENV infection induces strongly cross-neutralizing antibodies that protect against subsequent severe dengue disease. These immunologic interactions may be explained by differences in virion structure between DENV 1-4 and ZIKV, which modulate thermostability, susceptibility to neutralization, and cell infectivity. Overall, these observations are important for the understanding and prediction of epidemics and the development and evaluation of dengue and Zika vaccines.
Collapse
Affiliation(s)
- Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| |
Collapse
|
27
|
Zhao H, Xu L, Bombardi R, Nargi R, Deng Z, Errico JM, Nelson CA, Dowd KA, Pierson TC, Crowe JE, Diamond MS, Fremont DH. Mechanism of differential Zika and dengue virus neutralization by a public antibody lineage targeting the DIII lateral ridge. J Exp Med 2020; 217:jem.20191792. [PMID: 31757867 PMCID: PMC7041715 DOI: 10.1084/jem.20191792] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/09/2019] [Accepted: 10/18/2019] [Indexed: 12/20/2022] Open
Abstract
Evaluation of the human antibody response to Zika virus has identified common germline-derived mAbs capable of cross flavivirus neutralization. Zhao et al. provide a detailed mechanistic understanding of how flavivirus infections are prevented in a strain-specific manner by a representative mAb. We previously generated a panel of human monoclonal antibodies (mAbs) against Zika virus (ZIKV) and identified one, ZIKV-116, that shares germline usage with mAbs identified in multiple donors. Here we show that ZIKV-116 interferes with ZIKV infection at a post-cellular attachment step by blocking viral fusion with host membranes. ZIKV-116 recognizes the lateral ridge of envelope protein domain III, with one critical residue varying between the Asian and African strains responsible for differential binding affinity and neutralization potency (E393D). ZIKV-116 also binds to and cross-neutralizes some dengue virus serotype 1 (DENV1) strains, with genotype-dependent inhibition explained by variation in a domain II residue (R204K) that potentially modulates exposure of the distally located, partially cryptic epitope. The V-J reverted germline configuration of ZIKV-116 preferentially binds to and neutralizes an Asian ZIKV strain, suggesting that this epitope may optimally induce related B cell clonotypes. Overall, these studies provide a structural and molecular mechanism for a cross-reactive mAb that uniquely neutralizes ZIKV and DENV1.
Collapse
Affiliation(s)
- Haiyan Zhao
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Lily Xu
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Robin Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN
| | - Rachel Nargi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN
| | - Zengqin Deng
- Department of Cell Biology & Physiology, Washington University School of Medicine, Saint Louis, MO
| | - John M Errico
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Christopher A Nelson
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO
| | - Kimberly A Dowd
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - James E Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN.,Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO.,Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO.,Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO
| |
Collapse
|
28
|
Mapping the diverse structural landscape of the flavivirus antibody repertoire. Curr Opin Virol 2020; 45:51-64. [PMID: 32801077 DOI: 10.1016/j.coviro.2020.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 02/08/2023]
Abstract
Flaviviruses are emerging arthropod-borne RNA viruses, causing a broad spectrum of life-threatening disease symptoms such as encephalitis and hemorrhagic fever. Successful vaccines exist against yellow fever virus, Japanese encephalitis virus and tick-borne encephalitis virus. However, vaccine development against other flaviviruses like dengue virus is not straightforward. This is partly because of the high sequence conservation and immunological cross-reactivity among flavivirus envelope glycoproteins leading to antibody mediated enhancement of disease. A comprehensive analyses of the structural landscape of humoral immune response against flaviviruses is crucial for antigen design. Here, we compare the available structural data of several flavivirus antibody complexes with a major focus on Zika virus and dengue virus and discuss the mapped epitopes, the stoichiometry of antibody binding and mechanisms of neutralization.
Collapse
|
29
|
Carpp LN, Fong Y, Bonaparte M, Moodie Z, Juraska M, Huang Y, Price B, Zhuang Y, Shao J, Zheng L, Chambonneau L, Small R, Sridhar S, DiazGranados CA, Gilbert PB. Microneutralization assay titer correlates analysis in two phase 3 trials of the CYD-TDV tetravalent dengue vaccine in Asia and Latin America. PLoS One 2020; 15:e0234236. [PMID: 32542024 PMCID: PMC7295445 DOI: 10.1371/journal.pone.0234236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/07/2020] [Indexed: 12/26/2022] Open
Abstract
We previously showed that Month 13 50% plaque reduction neutralization test (PRNT50) neutralizing antibody (nAb) titers against dengue virus (DENV) correlated with vaccine efficacy (VE) of CYD-TDV against symptomatic, virologically-confirmed dengue (VCD) in the CYD14 and CYD15 Phase 3 trials. While PRNT is the gold standard nAb assay, it is time-consuming and costly. We developed a next-generation high-throughput microneutralization (MN) assay and assessed its suitability for immune-correlates analyses and immuno-bridging applications. We analyzed MN and PRNT50 titers measured at baseline and Month 13 in a randomly sampled immunogenicity subset, and at Month 13 in nearly all VCD cases through Month 25. For each serotype, MN and PRNT50 titers showed high correlations, at both baseline and Month 13, with MN yielding a higher frequency of baseline-seronegatives. For both assays, Month 13 titer correlated inversely with VCD risk. Like PRNT50, high Month 13 MN titers were associated with high VE, and estimated VE increased with average Month 13 MN titer. We also studied each assay as a valid surrogate endpoint based on the Prentice criteria, which supported each assay as a valid surrogate for DENV-1 but only partially valid for DENV-2, -3, and -4. In addition, we applied Super-Learner to assess how well demographic, Month 13 MN, and/or Month 13 PRNT50 titers could predict Month 13-25 VCD outcome status; prediction was best when using demographic, MN, and PRNT50 information. We conclude that Month 13 MN titer performs comparably to Month 13 PRNT50 titer as a correlate of risk, correlate of vaccine efficacy, and surrogate endpoint. The MN assay could potentially be used to assess nAb titers in immunogenicity studies, immune-correlates studies, and immuno-bridging applications. Additional research would be needed for assessing the utility of MN titer in correlates analyses of other DENV endpoints and over longer follow-up periods.
Collapse
Affiliation(s)
- Lindsay N. Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Matthew Bonaparte
- Global Clinical Immunology, Sanofi Pasteur, Swiftwater, Pennsylvania, United States of America
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Ying Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Brenda Price
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Yingying Zhuang
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Jason Shao
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Lingyi Zheng
- Global Clinical Immunology, Sanofi Pasteur, Swiftwater, Pennsylvania, United States of America
| | | | - Robert Small
- Sanofi Pasteur, Orlando, Florida, United States of America
| | | | | | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
30
|
Wilken L, Rimmelzwaan GF. Adaptive Immunity to Dengue Virus: Slippery Slope or Solid Ground for Rational Vaccine Design? Pathogens 2020; 9:pathogens9060470. [PMID: 32549226 PMCID: PMC7350362 DOI: 10.3390/pathogens9060470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/15/2022] Open
Abstract
The four serotypes of dengue virus are the most widespread causes of arboviral disease, currently placing half of the human population at risk of infection. Pre-existing immunity to one dengue virus serotype can predispose to severe disease following secondary infection with a different serotype. The phenomenon of immune enhancement has complicated vaccine development and likely explains the poor long-term safety profile of a recently licenced dengue vaccine. Therefore, alternative vaccine strategies should be considered. This review summarises studies dissecting the adaptive immune responses to dengue virus infection and (experimental) vaccination. In particular, we discuss the roles of (i) neutralising antibodies, (ii) antibodies to non-structural protein 1, and (iii) T cells in protection and pathogenesis. We also address how these findings could translate into next-generation vaccine approaches that mitigate the risk of enhanced dengue disease. Finally, we argue that the development of a safe and efficacious dengue vaccine is an attainable goal.
Collapse
|
31
|
Alaofi AL. Probing the flexibility of Zika virus envelope protein DIII epitopes using molecular dynamics simulations. MOLECULAR SIMULATION 2020. [DOI: 10.1080/08927022.2020.1738424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Ahmed L. Alaofi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
32
|
Barr KL, Schwarz ER, Prakoso D, Imtiaz K, Pu R, Morris JG, Khan E, Long MT. Strain-Dependent Activity of Zika Virus and Exposure History in Serological Diagnostics. Trop Med Infect Dis 2020; 5:tropicalmed5010038. [PMID: 32138262 PMCID: PMC7157670 DOI: 10.3390/tropicalmed5010038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Zika virus (ZIKV) circulates as two separate lineages, with significant genetic variability between strains. Strain-dependent activity has been reported for dengue virus, herpes simplex virus and influenza. Strain-dependent activity of subject specimens to a virus could be an impediment to serological diagnosis and vaccine development. In order to determine whether ZIKV exhibits strain-dependent activity when exposed to antibodies, we measured the neutralizing properties of polyclonal serum and three monoclonal antibodies (ZKA185, 753(3)C10, and 4G2) against three strains of ZIKV (MR−766, PRVABC59, and R103454). Here, MR−766 was inhibited almost 60% less by ZKA185 than PRVABC59 and R103454 (p = 0.008). ZKA185 enhanced dengue 4 infection up to 50% (p = 0.0058). PRVABC59 was not inhibited by mAb 753(3)C10 while MR−766 and R103453 were inhibited up to 90% (p = 0.04 and 0.036, respectively). Patient serum, regardless of exposure history, neutralized MR−766 ~30%−40% better than PRVABC56 or R103454 (p = 0.005−0.00007). The most troubling finding was the significant neutralization of MR−766 by patients with no ZIKV exposure. We also evaluated ZIKV antibody cross reactivity with various flaviviruses and found that more patients developed cross-reactive antibodies to Japanese encephalitis virus than the dengue viruses. The data here show that serological diagnosis of ZIKV is complicated and that qualitative neutralization assays cannot discriminate between flaviviruses.
Collapse
Affiliation(s)
- Kelli L. Barr
- Department of Biology, Baylor University, Waco, TX 76798, USA
- Correspondence:
| | - Erika R. Schwarz
- Department of Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (D.P.); (R.P.); (M.T.L.)
| | - Dhani Prakoso
- Department of Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (D.P.); (R.P.); (M.T.L.)
| | - Kehkashan Imtiaz
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi 74800, Pakistan; (K.I.); (E.K.)
| | - Ruiyu Pu
- Department of Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (D.P.); (R.P.); (M.T.L.)
| | - J. Glenn Morris
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32601, USA;
| | - Erum Khan
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi 74800, Pakistan; (K.I.); (E.K.)
| | - Maureen T. Long
- Department of Comparative Diagnostic and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (E.R.S.); (D.P.); (R.P.); (M.T.L.)
| |
Collapse
|
33
|
Hurtado-Monzón AM, Cordero-Rivera CD, Farfan-Morales CN, Osuna-Ramos JF, De Jesús-González LA, Reyes-Ruiz JM, Del Ángel RM. The role of anti-flavivirus humoral immune response in protection and pathogenesis. Rev Med Virol 2020; 30:e2100. [PMID: 32101633 DOI: 10.1002/rmv.2100] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Flavivirus infections are a public health threat in the world that requires the development of safe and effective vaccines. Therefore, the understanding of the anti-flavivirus humoral immune response is fundamental to future studies on flavivirus pathogenesis and the design of anti-flavivirus therapeutics. This review aims to provide an overview of the current understanding of the function and involvement of flavivirus proteins in the humoral immune response as well as the ability of the anti-envelope (anti-E) antibodies to interfere (neutralizing antibodies) or not (non-neutralizing antibodies) with viral infection, and how they can, in some circumstances enhance dengue virus infection on Fc gamma receptor (FcγR) bearing cells through a mechanism known as antibody-dependent enhancement (ADE). Thus, the dual role of the antibodies against E protein poses a formidable challenge for vaccine development. Also, we discuss the roles of antibody binding stoichiometry (the concentration, affinity, or epitope recognition) in the neutralization of flaviviruses and the "breathing" of flavivirus virions in the humoral immune response. Finally, the relevance of some specific antibodies in the design and improvement of effective vaccines is addressed.
Collapse
Affiliation(s)
- Arianna Mahely Hurtado-Monzón
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Daniel Cordero-Rivera
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Carlos Noe Farfan-Morales
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Juan Fidel Osuna-Ramos
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Luis Adrián De Jesús-González
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - José Manuel Reyes-Ruiz
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Rosa María Del Ángel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| |
Collapse
|
34
|
Flavivirus infection—A review of immunopathogenesis, immunological response, and immunodiagnosis. Virus Res 2019; 274:197770. [DOI: 10.1016/j.virusres.2019.197770] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022]
|
35
|
Zhang H, Kai ELJ, Lu L. Investigating the stability of dengue virus envelope protein dimer using well-tempered metadynamics simulations. Proteins 2019; 88:643-653. [PMID: 31697409 DOI: 10.1002/prot.25844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/24/2019] [Accepted: 11/03/2019] [Indexed: 12/21/2022]
Abstract
We explored the stability of the dengue virus envelope (E) protein dimer since it is widely assumed that the E protein dimer is stabilized by drug ligands or antibodies in an acidic environment, neutralizing the virus's ability to fuse with human cells. During this process, a large conformational change of the E protein dimer is required. We performed Molecular Dynamics simulations to mimic the conformational change and stability of the dimer in neutral and acidic conditions with the well-tempered metadynamics method. Furthermore, as a few neutralizing antibodies discovered from dengue patients were reported, we used the same simulation method to examine the influence of a selected antibody on the dimer stability in both neutral and acidic conditions. We also investigated the antibody's influence on a point-mutated E protein that had been reported to interrupt the protein-antibody interaction and result in more than 95% loss of the antibody's binding ability. Our simulation results are highly consistent with the experimental conclusion that binding of the antibody to the E protein dimer neutralizes the virus, especially in a low pH condition, while the mutation of W101A or N153A significantly reduces the antibody's ability in stabilizing the E protein dimer. We demonstrate that well-tempered metadynamics can be used to accurately explore the antibody's interaction on large protein complexes such as the E protein dimer, and the computational approach in this work is promising in future antibody development.
Collapse
Affiliation(s)
- Haiping Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Joint Engineering Research Center for Health Big Data Intelligent Analysis Technology, Shenzhen Institutes of Advanced Technology, Shenzhen, Guangdong Province, People's Republic of China
| | - Eric L J Kai
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lanyuan Lu
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
36
|
Earnest JT, Basore K, Roy V, Bailey AL, Wang D, Alter G, Fremont DH, Diamond MS. Neutralizing antibodies against Mayaro virus require Fc effector functions for protective activity. J Exp Med 2019; 216:2282-2301. [PMID: 31337735 PMCID: PMC6781005 DOI: 10.1084/jem.20190736] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/11/2019] [Accepted: 06/20/2019] [Indexed: 01/01/2023] Open
Abstract
Despite causing outbreaks of fever and arthritis in multiple countries, no countermeasures exist against Mayaro virus (MAYV), an emerging mosquito-transmitted alphavirus. We generated 18 neutralizing mAbs against MAYV, 11 of which had "elite" activity that inhibited infection with EC50 values of <10 ng/ml. Antibodies with the greatest inhibitory capacity in cell culture mapped to epitopes near the fusion peptide of E1 and in domain B of the E2 glycoproteins. Unexpectedly, many of the elite neutralizing mAbs failed to prevent MAYV infection and disease in vivo. Instead, the most protective mAbs bound viral antigen on the cell surface with high avidity and promoted specific Fc effector functions, including phagocytosis by neutrophils and monocytes. In subclass switching studies, murine IgG2a and humanized IgG1 mAb variants controlled infection better than murine IgG1 and humanized IgG1-N297Q variants. An optimally protective antibody response to MAYV and possibly other alphaviruses may require tandem virus neutralization by the Fab moiety and effector functions of the Fc region.
Collapse
Affiliation(s)
- James T Earnest
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Katherine Basore
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Adam L Bailey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - David Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
37
|
Faheem M, Barbosa Lima JC, Jamal SB, Silva PA, Barbosa JARG. An insight into dengue virus proteins as potential drug/vaccine targets. Future Virol 2019. [DOI: 10.2217/fvl-2019-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dengue virus (DENV) is an arbovirus that belongs to family flaviviridae. Its genome is composed of a single stranded RNA molecule that encodes a single polyprotein. The polyprotein is processed by viral and cellular proteases to generate ten viral proteins. There are four antigenically distinct serotypes of DENV (DENV1, DENV2, DENV3 and DENV4), which are genetically related. Although protein variability is a major problem in dengue treatment, the functional and structural studies of individual proteins are equally important in treatment development. The data accumulated on dengue proteins are significant to provide detailed understanding of viral infection, replication, host-immune evasion and pathogenesis. In this review, we summarized the detailed current knowledge about DENV proteins.
Collapse
Affiliation(s)
- Muhammad Faheem
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| | - Jônatas Cunha Barbosa Lima
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
| | - Syed Babar Jamal
- Department of Biological Sciences, National University of Medical Sciences, The Mall road, Rawalpindi, Punjab 46000, Pakistan
| | - Paula Andreia Silva
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| | - João Alexandre Ribeiro Gonçalves Barbosa
- Laboratory of Biophysics, Department of Cellular Biology, University of Brasilia, Brasilia-DF 70910-900, Brazil
- Post-graduate program of Genomics Sciences & Biotechnology, Catholic University of Brasilia, Brasília-DF 70790-160, Brazil
| |
Collapse
|
38
|
Dowd KA, Pierson TC. The Many Faces of a Dynamic Virion: Implications of Viral Breathing on Flavivirus Biology and Immunogenicity. Annu Rev Virol 2019; 5:185-207. [PMID: 30265634 DOI: 10.1146/annurev-virology-092917-043300] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Flaviviruses are arthropod-borne RNA viruses that are a significant threat to global health due to their widespread distribution, ability to cause severe disease in humans, and capacity for explosive spread following introduction into new regions. Members of this genus include dengue, tick-borne encephalitis, yellow fever, and Zika viruses. Vaccination has been a highly successful means to control flaviviruses, and neutralizing antibodies are an important component of a protective immune response. High-resolution structures of flavivirus structural proteins and virions, alone and in complex with antibodies, provide a detailed understanding of viral fusion mechanisms and virus-antibody interactions. However, mounting evidence suggests these structures provide only a snapshot of an otherwise structurally dynamic virus particle. The contribution of the structural ensemble arising from viral breathing to the biology, antigenicity, and immunity of flaviviruses is discussed, including implications for the development and evaluation of flavivirus vaccines.
Collapse
Affiliation(s)
- Kimberly A Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| | - Theodore C Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| |
Collapse
|
39
|
Keasey SL, Smith JL, Fernandez S, Durbin AP, Zhao BM, Ulrich RG. Impact of Dengue Virus Serotype 2 Strain Diversity on Serological Immune Responses to Dengue. ACS Infect Dis 2018; 4:1705-1717. [PMID: 30347144 DOI: 10.1021/acsinfecdis.8b00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dengue is a mosquito-borne disease caused by four dengue virus serotypes (DENV1-4) that are loosely categorized by sequence commonalities and antibody recognition profiles. The highly variable envelope protein (E) that is prominently displayed on the surface of DENV is an essential component of vaccines currently under development, yet the impact of using single strains to represent each serotype in tetravalent vaccines has not been adequately studied. We synthesized chimeric E by replacing highly variable residues from a dengue virus serotype 2 vaccine strain (PUO-218) with those from 16 DENV2 lineages spanning 60 years of antigen evolution. Examining sera from human and rhesus macaques challenged with single strains of DENV2, antibody-E interactions were markedly inhibited or enhanced by residues mainly focused within a 480 Å2 footprint displayed on the E backbone. The striking impact of E diversity on polyclonal immune responses suggests that frequent antigen updates may be necessary for vaccines to counter shifts in circulating strains.
Collapse
Affiliation(s)
- Sarah L. Keasey
- Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, Maryland 21702, United States
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, Maryland 21250, United States
| | - Jessica L. Smith
- Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, Maryland 21702, United States
| | - Stefan Fernandez
- Armed Forces Research Institute of Medical Sciences, Bangkok, 10400, Thailand
| | - Anna P. Durbin
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 624 North Broadway, Room 251, Baltimore, Maryland 21205, United States
| | - Bryan M. Zhao
- Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, Maryland 21702, United States
| | - Robert G. Ulrich
- Molecular and Translational Sciences Division, U.S. Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Frederick, Maryland 21702, United States
| |
Collapse
|
40
|
Characterization of a potent and highly unusual minimally enhancing antibody directed against dengue virus. Nat Immunol 2018; 19:1248-1256. [PMID: 30323338 DOI: 10.1038/s41590-018-0227-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 08/20/2018] [Indexed: 12/12/2022]
Abstract
Dengue virus is a major pathogen, and severe infections can lead to life-threatening dengue hemorrhagic fever. Dengue virus exists as four serotypes, and dengue hemorrhagic fever is often associated with secondary heterologous infections. Antibody-dependent enhancement (ADE) may drive higher viral loads in these secondary infections and is purported to result from antibodies that recognize dengue virus but fail to fully neutralize it. Here we characterize two antibodies, 2C8 and 3H5, that bind to the envelope protein. Antibody 3H5 is highly unusual as it not only is potently neutralizing but also promotes little if any ADE, whereas antibody 2C8 has strong capacity to promote ADE. We show that 3H5 shows resilient binding in endosomal pH conditions and neutralizes at low occupancy. Immunocomplexes of 3H5 and dengue virus do not efficiently interact with Fcγ receptors, which we propose is due to the binding mode of 3H5 and constitutes the primary mechanism of how ADE is avoided.
Collapse
|
41
|
Barrows NJ, Campos RK, Liao KC, Prasanth KR, Soto-Acosta R, Yeh SC, Schott-Lerner G, Pompon J, Sessions OM, Bradrick SS, Garcia-Blanco MA. Biochemistry and Molecular Biology of Flaviviruses. Chem Rev 2018; 118:4448-4482. [PMID: 29652486 DOI: 10.1021/acs.chemrev.7b00719] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Flaviviruses, such as dengue, Japanese encephalitis, tick-borne encephalitis, West Nile, yellow fever, and Zika viruses, are critically important human pathogens that sicken a staggeringly high number of humans every year. Most of these pathogens are transmitted by mosquitos, and not surprisingly, as the earth warms and human populations grow and move, their geographic reach is increasing. Flaviviruses are simple RNA-protein machines that carry out protein synthesis, genome replication, and virion packaging in close association with cellular lipid membranes. In this review, we examine the molecular biology of flaviviruses touching on the structure and function of viral components and how these interact with host factors. The latter are functionally divided into pro-viral and antiviral factors, both of which, not surprisingly, include many RNA binding proteins. In the interface between the virus and the hosts we highlight the role of a noncoding RNA produced by flaviviruses to impair antiviral host immune responses. Throughout the review, we highlight areas of intense investigation, or a need for it, and potential targets and tools to consider in the important battle against pathogenic flaviviruses.
Collapse
Affiliation(s)
- Nicholas J Barrows
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Department of Molecular Genetics and Microbiology , Duke University , Durham , North Carolina 27710 , United States
| | - Rafael K Campos
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Department of Molecular Genetics and Microbiology , Duke University , Durham , North Carolina 27710 , United States
| | - Kuo-Chieh Liao
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - K Reddisiva Prasanth
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Ruben Soto-Acosta
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Shih-Chia Yeh
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - Geraldine Schott-Lerner
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Julien Pompon
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore.,MIVEGEC, IRD, CNRS, Université de Montpellier , Montpellier 34090 , France
| | - October M Sessions
- Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , Texas 77555 , United States.,Programme in Emerging Infectious Diseases , Duke-NUS Medical School , Singapore 169857 , Singapore
| |
Collapse
|
42
|
Fernandez E, Kose N, Edeling MA, Adhikari J, Sapparapu G, Lazarte SM, Nelson CA, Govero J, Gross ML, Fremont DH, Crowe JE, Diamond MS. Mouse and Human Monoclonal Antibodies Protect against Infection by Multiple Genotypes of Japanese Encephalitis Virus. mBio 2018; 9:e00008-18. [PMID: 29487230 PMCID: PMC5829823 DOI: 10.1128/mbio.00008-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 01/26/2018] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis virus (JEV) remains a leading cause of viral encephalitis worldwide. Although JEV-specific antibodies have been described, an assessment of their ability to neutralize multiple genotypes of JEV has been limited. Here, we describe the development of a panel of mouse and human neutralizing monoclonal antibodies (MAbs) that inhibit infection in cell culture of four different JEV genotypes tested. Mechanism-of-action studies showed that many of these MAbs inhibited infection at a postattachment step, including blockade of virus fusion. Mapping studies using site-directed mutagenesis and hydrogen-deuterium exchange with mass spectrometry revealed that the lateral ridge on domain III of the envelope protein was a primary recognition epitope for our panel of strongly neutralizing MAbs. Therapeutic studies in mice demonstrated protection against lethality caused by genotype I and III strains when MAbs were administered as a single dose even 5 days after infection. This information may inform the development of vaccines and therapeutic antibodies as emerging strains and genotypic shifts become more prevalent.IMPORTANCE Although Japanese encephalitis virus (JEV) is a vaccine-preventable cause of viral encephalitis, the inactivated and live attenuated platforms available are derived from strains belonging to a single genotype (GIII) due to its historical prevalence in areas of JEV epidemics. Related to this, studies with vaccines and antibodies have focused on assessing the in vitro and in vivo protective responses to homologous or heterologous GIII strains. An epidemiological shift in JEV genotype distribution warrants the induction of broadly neutralizing antibody responses that inhibit infection of multiple JEV genotypes. Here, we generated a panel of mouse and human neutralizing monoclonal antibodies and evaluated their inhibitory activity, epitope location, and capacity for protection against multiple JEV genotypes in mice.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/immunology
- Chlorocebus aethiops
- Disease Models, Animal
- Encephalitis Virus, Japanese/classification
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Epitopes/immunology
- Genotype
- Humans
- Mice
- Models, Biological
- Treatment Outcome
- Vero Cells
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Nurgun Kose
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Melissa A Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jagat Adhikari
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Gopal Sapparapu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susana M Lazarte
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jennifer Govero
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
43
|
Development of Envelope Protein Antigens To Serologically Differentiate Zika Virus Infection from Dengue Virus Infection. J Clin Microbiol 2018; 56:JCM.01504-17. [PMID: 29263206 DOI: 10.1128/jcm.01504-17] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/11/2017] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that can cause birth defects and neurologic complications. Molecular tests are effective for diagnosing acute ZIKV infection, although the majority of infections produce no symptoms at all or present after the narrow window in which molecular diagnostics are dependable. Serology is a reliable method for detecting infections after the viremic period; however, most serological assays have limited specificity due to cross-reactive antibodies elicited by flavivirus infections. Since ZIKV and dengue virus (DENV) widely cocirculate, distinguishing ZIKV infection from DENV infection is particularly important for diagnosing individual cases or for surveillance to coordinate public health responses. Flaviviruses also elicit type-specific antibodies directed to non-cross-reactive epitopes of the infecting virus; such epitopes are attractive targets for the design of antigens for development of serological tests with greater specificity. Guided by comparative epitope modeling of the ZIKV envelope protein, we designed two recombinant antigens displaying unique antigenic regions on domain I (Z-EDI) and domain III (Z-EDIII) of the ZIKV envelope protein. Both the Z-EDI and Z-EDIII antigens consistently detected ZIKV-specific IgG in ZIKV-immune sera but not cross-reactive IgG in DENV-immune sera in late convalescence (>12 weeks postinfection). In contrast, during early convalescence (2 to 12 weeks postinfection), secondary DENV-immune sera and some primary DENV-immune sera cross-reacted with the Z-EDI and Z-EDIII antigens. Analysis of sequential samples from DENV-immune individuals demonstrated that Z-EDIII cross-reactivity peaked in early convalescence and declined steeply over time. The Z-EDIII antigen has much potential as a diagnostic antigen for population-level surveillance and for detecting past infections in patients.
Collapse
|
44
|
Rey FA, Stiasny K, Vaney MC, Dellarole M, Heinz FX. The bright and the dark side of human antibody responses to flaviviruses: lessons for vaccine design. EMBO Rep 2018; 19:206-224. [PMID: 29282215 PMCID: PMC5797954 DOI: 10.15252/embr.201745302] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 01/07/2023] Open
Abstract
Zika and dengue viruses belong to the Flavivirus genus, a close group of antigenically related viruses that cause significant arthropod-transmitted diseases throughout the globe. Although infection by a given flavivirus is thought to confer lifelong protection, some of the patient's antibodies cross-react with other flaviviruses without cross-neutralizing. The original antigenic sin phenomenon may amplify such antibodies upon subsequent heterologous flavivirus infection, potentially aggravating disease by antibody-dependent enhancement (ADE). The most striking example is provided by the four different dengue viruses, where infection by one serotype appears to predispose to more severe disease upon infection by a second one. A similar effect was postulated for sequential infections with Zika and dengue viruses. In this review, we analyze the molecular determinants of the dual antibody response to flavivirus infection or vaccination in humans. We highlight the role of conserved partially cryptic epitopes giving rise to cross-reacting and poorly neutralizing, ADE-prone antibodies. We end by proposing a strategy for developing an epitope-focused vaccine approach to avoid eliciting undesirable antibodies while focusing the immune system on producing protective antibodies only.
Collapse
Affiliation(s)
- Félix A Rey
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Marie-Christine Vaney
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Mariano Dellarole
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Franz X Heinz
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
45
|
Goo L, DeMaso CR, Pelc RS, Ledgerwood JE, Graham BS, Kuhn RJ, Pierson TC. The Zika virus envelope protein glycan loop regulates virion antigenicity. Virology 2018; 515:191-202. [PMID: 29304471 DOI: 10.1016/j.virol.2017.12.032] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 12/22/2017] [Accepted: 12/26/2017] [Indexed: 01/30/2023]
Abstract
Because antibodies are an important component of flavivirus immunity, understanding the antigenic structure of flaviviruses is critical. Compared to dengue virus (DENV), the loop containing the single N-linked glycosylation site on Zika virus (ZIKV) envelope (E) proteins extends further towards the DII fusion loop (DII-FL) on neighboring E proteins within E dimers on mature viruses. Although ZIKV is poorly neutralized by DII-FL antibodies, we demonstrated significantly increased neutralization sensitivity of ZIKV particles incorporating the DENV glycan loop. Increased neutralization sensitivity was independent of E protein glycosylation: ZIKV lacking E protein glycans remained poorly neutralized, whereas ZIKV loop chimeras with or without an E protein glycan were potently neutralized. ZIKV particles lacking the E protein glycan were capable of infecting Raji cells expressing the lectin DC-SIGNR, suggesting the prM glycan of partially mature particles can facilitate entry. Our study provides insight into the determinants of ZIKV E protein function and antigenicity.
Collapse
Affiliation(s)
- Leslie Goo
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Christina R DeMaso
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Rebecca S Pelc
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Julie E Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard J Kuhn
- Markey Center for Structural Biology and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America.
| |
Collapse
|
46
|
Development of Antibody Therapeutics against Flaviviruses. Int J Mol Sci 2017; 19:ijms19010054. [PMID: 29295568 PMCID: PMC5796004 DOI: 10.3390/ijms19010054] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/28/2022] Open
Abstract
Recent outbreaks of Zika virus (ZIKV) highlight the urgent need to develop efficacious interventions against flaviviruses, many of which cause devastating epidemics around the world. Monoclonal antibodies (mAb) have been at the forefront of treatment for cancer and a wide array of other diseases due to their specificity and potency. While mammalian cell-produced mAbs have shown promise as therapeutic candidates against several flaviviruses, their eventual approval for human application still faces several challenges including their potential risk of predisposing treated patients to more severe secondary infection by a heterologous flavivirus through antibody-dependent enhancement (ADE). The high cost associated with mAb production in mammalian cell cultures also poses a challenge for the feasible application of these drugs to the developing world where the majority of flavivirus infection occurs. Here, we review the current therapeutic mAb candidates against various flaviviruses including West Nile (WNV), Dengue virus (DENV), and ZIKV. The progress of using plants for developing safer and more economical mAb therapeutics against flaviviruses is discussed within the context of their expression, characterization, downstream processing, neutralization, and in vivo efficacy. The progress of using plant glycoengineering to address ADE, the major impediment of flavivirus therapeutic development, is highlighted. These advancements suggest that plant-based systems are excellent alternatives for addressing the remaining challenges of mAb therapeutic development against flavivirus and may facilitate the eventual commercialization of these drug candidates.
Collapse
|
47
|
Li J, Watterson D, Chang CW, Che XY, Li XQ, Ericsson DJ, Qiu LW, Cai JP, Chen J, Fry SR, Cheung STM, Cooper MA, Young PR, Kobe B. Structural and Functional Characterization of a Cross-Reactive Dengue Virus Neutralizing Antibody that Recognizes a Cryptic Epitope. Structure 2017; 26:51-59.e4. [PMID: 29249606 DOI: 10.1016/j.str.2017.11.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/08/2017] [Accepted: 11/17/2017] [Indexed: 10/18/2022]
Abstract
Understanding the molecular basis of the neutralizing antibody response to dengue virus (DENV) is an essential component in the design and development of effective vaccines and immunotherapeutics. Here we present the structure of a cross-reactive, neutralizing antibody, 3E31, in complex with domain III (DIII) of the DENV envelope (E) protein and reveal a conserved, temperature-sensitive, cryptic epitope on DIII that is not available in any of the known conformations of E on the dengue virion. We observed that 3E31 inhibits E-mediated membrane fusion, suggesting that the antibody is able to neutralize virus through binding an as-yet uncharacterized intermediate conformation of DENV E and sterically block trimer formation. Finally, we show that, unlike cross-reactive fusion peptide-specific antibodies, 3E31 does not promote antibody-dependent enhancement of infection at sub-neutralizing concentrations. Our results highlight the 3E31 epitope on the E protein DIII as a promising target for immunotherapeutics or vaccine design.
Collapse
Affiliation(s)
- Jie Li
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China; School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Chiung-Wen Chang
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiao-Yan Che
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Xiao-Quan Li
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Daniel J Ericsson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia
| | - Li-Wen Qiu
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Jian-Piao Cai
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Jing Chen
- Division of Laboratory Medicine, ZhuJiang Hospital, Southern Medical University, Guangzhou 510282, People's Republic of China
| | - Scott R Fry
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia
| | - Stacey T M Cheung
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Matthew A Cooper
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
48
|
Katzelnick LC, Gresh L, Halloran ME, Mercado JC, Kuan G, Gordon A, Balmaseda A, Harris E. Antibody-dependent enhancement of severe dengue disease in humans. Science 2017; 358:929-932. [PMID: 29097492 PMCID: PMC5858873 DOI: 10.1126/science.aan6836] [Citation(s) in RCA: 782] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Accepted: 09/29/2017] [Indexed: 01/09/2023]
Abstract
For dengue viruses 1 to 4 (DENV1-4), a specific range of antibody titer has been shown to enhance viral replication in vitro and severe disease in animal models. Although suspected, such antibody-dependent enhancement of severe disease has not been shown to occur in humans. Using multiple statistical approaches to study a long-term pediatric cohort in Nicaragua, we show that risk of severe dengue disease is highest within a narrow range of preexisting anti-DENV antibody titers. By contrast, we observe protection from all symptomatic dengue disease at high antibody titers. Thus, immune correlates of severe dengue must be evaluated separately from correlates of protection against symptomatic disease. These results have implications for studies of dengue pathogenesis and for vaccine development, because enhancement, not just lack of protection, is of concern.
Collapse
Affiliation(s)
- Leah C. Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| | - Lionel Gresh
- Sustainable Sciences Institute, Managua, Nicaragua
| | - M. Elizabeth Halloran
- Department of Biostatistics, University of Washington, WA, USA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Juan Carlos Mercado
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Guillermina Kuan
- Centro de Salud Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
| | - Aubree Gordon
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Angel Balmaseda
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministry of Health, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, CA, USA
| |
Collapse
|
49
|
Clark MJ, Miduturu C, Schmidt AG, Zhu X, Pitts JD, Wang J, Potisopon S, Zhang J, Wojciechowski A, Hann Chu JJ, Gray NS, Yang PL. GNF-2 Inhibits Dengue Virus by Targeting Abl Kinases and the Viral E Protein. Cell Chem Biol 2017; 23:443-52. [PMID: 27105280 DOI: 10.1016/j.chembiol.2016.03.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 03/12/2016] [Accepted: 03/16/2016] [Indexed: 12/11/2022]
Abstract
Dengue virus infects more than 300 million people annually, yet there is no widely protective vaccine or drugs against the virus. Efforts to develop antivirals against classical targets such as the viral protease and polymerase have not yielded drugs that have advanced to the clinic. Here, we show that the allosteric Abl kinase inhibitor GNF-2 interferes with dengue virus replication via activity mediated by cellular Abl kinases but additionally blocks viral entry via an Abl-independent mechanism. To characterize this newly discovered antiviral activity, we developed disubstituted pyrimidines that block dengue virus entry with structure-activity relationships distinct from those driving kinase inhibition. We demonstrate that biotin- and fluorophore-conjugated derivatives of GNF-2 interact with the dengue glycoprotein, E, in the pre-fusion conformation that exists on the virion surface, and that this interaction inhibits viral entry. This study establishes GNF-2 as an antiviral compound with polypharmacological activity and provides "lead" compounds for further optimization efforts.
Collapse
Affiliation(s)
- Margaret J Clark
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Chandra Miduturu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Avenue, Longwood Center LC-2209, Boston, MA 02115, USA
| | - Aaron G Schmidt
- Laboratory of Molecular Medicine, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xuling Zhu
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jared D Pitts
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Avenue, Longwood Center LC-2209, Boston, MA 02115, USA
| | - Supanee Potisopon
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jianming Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Avenue, Longwood Center LC-2209, Boston, MA 02115, USA
| | - Amy Wojciechowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Avenue, Longwood Center LC-2209, Boston, MA 02115, USA
| | - Justin Jang Hann Chu
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biological Chemistry & Molecular Pharmacology, Harvard Medical School, 360 Longwood Avenue, Longwood Center LC-2209, Boston, MA 02115, USA
| | - Priscilla L Yang
- Department of Microbiology and Immunobiology, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| |
Collapse
|
50
|
Wu Y, Li S, Du L, Wang C, Zou P, Hong B, Yuan M, Ren X, Tai W, Kong Y, Zhou C, Lu L, Zhou X, Jiang S, Ying T. Neutralization of Zika virus by germline-like human monoclonal antibodies targeting cryptic epitopes on envelope domain III. Emerg Microbes Infect 2017; 6:e89. [PMID: 29018252 PMCID: PMC5658772 DOI: 10.1038/emi.2017.79] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 08/01/2017] [Accepted: 08/06/2017] [Indexed: 12/11/2022]
Abstract
The Zika virus (ZIKV), a flavivirus transmitted by Aedes mosquitoes, has emerged as a global public health concern. Pre-existing cross-reactive antibodies against other flaviviruses could modulate immune responses to ZIKV infection by antibody-dependent enhancement, highlighting the importance of understanding the immunogenicity of the ZIKV envelope protein. In this study, we identified a panel of human monoclonal antibodies (mAbs) that target domain III (DIII) of the ZIKV envelope protein from a very large phage-display naive antibody library. These germline-like antibodies, sharing 98%-100% hoLogy with their corresponding germline IGHV genes, bound ZIKV DIII specifically with high affinities. One mAb, m301, broadly neutralized the currently circulating ZIKV strains and showed a synergistic effect with another mAb, m302, in neutralizing ZIKV in vitro and in a mouse model of ZIKV infection. Interestingly, epitope mapping and competitive binding studies suggest that m301 and m302 bind adjacent regions of the DIII C-C' loop, which represents a recently identified cryptic epitope that is intermittently exposed in an uncharacterized virus conformation. This study extended our understanding of antigenic epitopes of ZIKV antibodies and has direct implications for the design of ZIKV vaccines.
Collapse
Affiliation(s)
- Yanling Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Shun Li
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lanying Du
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Chunyu Wang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Binbin Hong
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Mengjiao Yuan
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaonan Ren
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Wanbo Tai
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Yu Kong
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Chen Zhou
- Biomissile Corporation, Shanghai 201203, China
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|