1
|
Boggiano-Ayo T, Palacios-Oliva J, Lozada-Chang S, Relova-Hernandez E, Gomez-Perez J, Oliva G, Hernandez L, Bueno-Soler A, Montes de Oca D, Mora O, Machado-Santisteban R, Perez-Martinez D, Perez-Masson B, Cabrera Infante Y, Calzadilla-Rosado L, Ramirez Y, Aymed-Garcia J, Ruiz-Ramirez I, Romero Y, Gomez T, Espinosa LA, Gonzalez LJ, Cabrales A, Guirola O, de la Luz KR, Pi-Estopiñan F, Sanchez-Ramirez B, Garcia-Rivera D, Valdes-Balbin Y, Rojas G, Leon-Monzon K, Ojito-Magaz E, Hardy E. Development of a scalable single process for producing SARS-CoV-2 RBD monomer and dimer vaccine antigens. Front Bioeng Biotechnol 2023; 11:1287551. [PMID: 38050488 PMCID: PMC10693982 DOI: 10.3389/fbioe.2023.1287551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/30/2023] [Indexed: 12/06/2023] Open
Abstract
We have developed a single process for producing two key COVID-19 vaccine antigens: SARS-CoV-2 receptor binding domain (RBD) monomer and dimer. These antigens are featured in various COVID-19 vaccine formats, including SOBERANA 01 and the licensed SOBERANA 02, and SOBERANA Plus. Our approach involves expressing RBD (319-541)-His6 in Chinese hamster ovary (CHO)-K1 cells, generating and characterizing oligoclones, and selecting the best RBD-producing clones. Critical parameters such as copper supplementation in the culture medium and cell viability influenced the yield of RBD dimer. The purification of RBD involved standard immobilized metal ion affinity chromatography (IMAC), ion exchange chromatography, and size exclusion chromatography. Our findings suggest that copper can improve IMAC performance. Efficient RBD production was achieved using small-scale bioreactor cell culture (2 L). The two RBD forms - monomeric and dimeric RBD - were also produced on a large scale (500 L). This study represents the first large-scale application of perfusion culture for the production of RBD antigens. We conducted a thorough analysis of the purified RBD antigens, which encompassed primary structure, protein integrity, N-glycosylation, size, purity, secondary and tertiary structures, isoform composition, hydrophobicity, and long-term stability. Additionally, we investigated RBD-ACE2 interactions, in vitro ACE2 recognition of RBD, and the immunogenicity of RBD antigens in mice. We have determined that both the monomeric and dimeric RBD antigens possess the necessary quality attributes for vaccine production. By enabling the customizable production of both RBD forms, this unified manufacturing process provides the required flexibility to adapt rapidly to the ever-changing demands of emerging SARS-CoV-2 variants and different COVID-19 vaccine platforms.
Collapse
Affiliation(s)
- Tammy Boggiano-Ayo
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | | | | | - Gonzalo Oliva
- Process Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Alexi Bueno-Soler
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Osvaldo Mora
- Process Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Dayana Perez-Martinez
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Beatriz Perez-Masson
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | - Yaima Ramirez
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Judey Aymed-Garcia
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Yamile Romero
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Tania Gomez
- Quality Direction, Center of Molecular Immunology, Havana, Cuba
| | | | | | - Annia Cabrales
- Center for Genetic Engineering and Biotechnology, Playa, Cuba
| | - Osmany Guirola
- Center for Genetic Engineering and Biotechnology, Playa, Cuba
| | | | | | | | | | | | - Gertrudis Rojas
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | - Kalet Leon-Monzon
- Immunology and Immunobiology Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Eugenio Hardy
- Process Development Direction, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
2
|
Meganck RM, Zhu D, Dong S, Snoderly-Foster LJ, Dalben YR, Thiono D, White LJ, DeSilva AM, Baric RS, Tse LV. Evolution of a functionally intact but antigenically distinct DENV fusion loop. eLife 2023; 12:RP87555. [PMID: 37725085 PMCID: PMC10508882 DOI: 10.7554/elife.87555] [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: 09/21/2023] Open
Abstract
A hallmark of dengue virus (DENV) pathogenesis is the potential for antibody-dependent enhancement, which is associated with deadly DENV secondary infection, complicates the identification of correlates of protection, and negatively impacts the safety and efficacy of DENV vaccines. Antibody-dependent enhancement is linked to antibodies targeting the fusion loop (FL) motif of the envelope protein, which is completely conserved in mosquito-borne flaviviruses and required for viral entry and fusion. In the current study, we utilized saturation mutagenesis and directed evolution to engineer a functional variant with a mutated FL (D2-FL), which is not neutralized by FL-targeting monoclonal antibodies. The FL mutations were combined with our previously evolved prM cleavage site to create a mature version of D2-FL (D2-FLM), which evades both prM- and FL-Abs but retains sensitivity to other type-specific and quaternary cross-reactive (CR) Abs. CR serum from heterotypic (DENV4)-infected non-human primates (NHP) showed lower neutralization titers against D2-FL and D2-FLM than isogenic wildtype DENV2 while similar neutralization titers were observed in serum from homotypic (DENV2)-infected NHP. We propose D2-FL and D2-FLM as valuable tools to delineate CR Ab subtypes in serum as well as an exciting platform for safer live-attenuated DENV vaccines suitable for naïve individuals and children.
Collapse
Affiliation(s)
- Rita M Meganck
- Department of Molecular Microbiology and Immunology, Saint Louis UniversitySaint LouisUnited States
| | - Deanna Zhu
- Department of Epidemiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Lisa J Snoderly-Foster
- Department of Molecular Microbiology and Immunology, Saint Louis UniversitySaint LouisUnited States
| | - Yago R Dalben
- Department of Molecular Microbiology and Immunology, Saint Louis UniversitySaint LouisUnited States
| | - Devina Thiono
- Department of Microbiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Laura J White
- Department of Microbiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Arivianda M DeSilva
- Department of Microbiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel HillChapel HillUnited States
| | - Longping V Tse
- Department of Molecular Microbiology and Immunology, Saint Louis UniversitySaint LouisUnited States
| |
Collapse
|
3
|
Meganck RM, Zhu D, Dong S, Snoderly-Foster LJ, Dalben YR, Thiono D, White LJ, DeSilva AM, Baric RS, Tse LV. Evolution of a Functionally Intact but Antigenically Distinct DENV Fusion Loop. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.22.533803. [PMID: 37034784 PMCID: PMC10081285 DOI: 10.1101/2023.03.22.533803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A hallmark of Dengue virus (DENV) pathogenesis is the potential for antibody-dependent enhancement, which is associated with deadly DENV secondary infection, complicates the identification of correlates of protection, and negatively impacts the safety and efficacy of DENV vaccines. ADE is linked to antibodies targeting the fusion loop (FL) motif of the envelope protein, which is completely conserved in mosquito-borne flaviviruses and required for viral entry and fusion. In the current study, we utilized saturation mutagenesis and directed evolution to engineer a functional variant with a mutated FL (D2-FL) which is not neutralized by FL-targeting monoclonal antibodies. The FL mutations were combined with our previously evolved prM cleavage site to create a mature version of D2-FL (D2-FLM), which evades both prM- and FL-Abs but retains sensitivity to other type-specific and quaternary cross-reactive (CR) Abs. CR serum from heterotypic (DENV4) infected non-human primates (NHP) showed lower neutralization titers against D2-FL and D2-FLM than isogenic wildtype DENV2 while similar neutralization titers were observed in serum from homotypic (DENV2) infected NHP. We propose D2-FL and D2-FLM as valuable tools to delineate CR Ab subtypes in serum as well as an exciting platform for safer live attenuated DENV vaccines suitable for naïve individuals and children.
Collapse
Affiliation(s)
- Rita M. Meganck
- Department of Molecular Microbiology and Immunology, Saint Louis University
| | - Deanna Zhu
- Department of Epidemiology, University of North Carolina at Chapel Hill
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina at Chapel Hill
| | | | - Yago R. Dalben
- Department of Molecular Microbiology and Immunology, Saint Louis University
| | - Devina Thiono
- Department of Microbiology, University of North Carolina at Chapel Hill
| | - Laura J. White
- Department of Microbiology, University of North Carolina at Chapel Hill
| | | | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill
| | - Longping V. Tse
- Department of Molecular Microbiology and Immunology, Saint Louis University
| |
Collapse
|
4
|
Adams C, Carbaugh DL, Shu B, Ng TS, Castillo IN, Bhowmik R, Segovia-Chumbez B, Puhl AC, Graham S, Diehl SA, Lazear HM, Lok SM, de Silva AM, Premkumar L. Structure and neutralization mechanism of a human antibody targeting a complex Epitope on Zika virus. PLoS Pathog 2023; 19:e1010814. [PMID: 36626401 PMCID: PMC9870165 DOI: 10.1371/journal.ppat.1010814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 01/23/2023] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
We currently have an incomplete understanding of why only a fraction of human antibodies that bind to flaviviruses block infection of cells. Here we define the footprint of a strongly neutralizing human monoclonal antibody (mAb G9E) with Zika virus (ZIKV) by both X-ray crystallography and cryo-electron microscopy. Flavivirus envelope (E) glycoproteins are present as homodimers on the virion surface, and G9E bound to a quaternary structure epitope spanning both E protomers forming a homodimer. As G9E mainly neutralized ZIKV by blocking a step after viral attachment to cells, we tested if the neutralization mechanism of G9E was dependent on the mAb cross-linking E molecules and blocking low-pH triggered conformational changes required for viral membrane fusion. We introduced targeted mutations to the G9E paratope to create recombinant antibodies that bound to the ZIKV envelope without cross-linking E protomers. The G9E paratope mutants that bound to a restricted epitope on one protomer poorly neutralized ZIKV compared to the wild-type mAb, demonstrating that the neutralization mechanism depended on the ability of G9E to cross-link E proteins. In cell-free low pH triggered viral fusion assay, both wild-type G9E, and epitope restricted paratope mutant G9E bound to ZIKV but only the wild-type G9E blocked fusion. We propose that, beyond antibody binding strength, the ability of human antibodies to cross-link E-proteins is a critical determinant of flavivirus neutralization potency.
Collapse
Affiliation(s)
- Cameron Adams
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Derek L. Carbaugh
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Bo Shu
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Centre for Bio-Imaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Thiam-Seng Ng
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Centre for Bio-Imaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Izabella N. Castillo
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Ryan Bhowmik
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Bruno Segovia-Chumbez
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Ana C. Puhl
- Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Stephen Graham
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Sean A. Diehl
- Department of Microbiology and Molecular Genetics, University of Vermont Larner College of Medicine, Burlington, Vermont, United States of America
| | - Helen M. Lazear
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Shee-mei Lok
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Centre for Bio-Imaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
5
|
Generation of soluble, cleaved, well-ordered, native-like dimers of dengue virus 4 envelope protein ectodomain (sE) suitable for vaccine immunogen design. Int J Biol Macromol 2022; 217:19-26. [PMID: 35817240 DOI: 10.1016/j.ijbiomac.2022.07.028] [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: 09/03/2021] [Revised: 11/12/2021] [Accepted: 07/04/2022] [Indexed: 11/24/2022]
Abstract
Dengue virus is transmitted by Aedes mosquitoes and dengue is endemic in many regions of the world. Severe dengue results in complications that may lead to death. Although some vaccine candidates are in clinical trials and one vaccine Dengvaxia, with restricted efficacy, is available, there are currently no specific therapies to completely prevent or treat dengue. The dengue virus structural protein E (envelope) exists as a head-to-tail dimer on mature virus, is targeted by broadly neutralizing antibodies and is suitable for developing vaccine immunogens. Here, we have used a redesigned dengue prME expression construct and immunoaffinity chromatography with conformational/quaternary antibody A11 to purify soluble DENV4 sE(A259C) (E ectodomain) dimers from mammalian expression system to ~99 % purity. These dimers retain glycosylation reported for native DENV E, display the three major broadly neutralizing antibody epitopes, and form well-ordered structure. This strategy can be used for developing subunit vaccine candidates against dengue and other flaviviruses.
Collapse
|
6
|
Thompson D, Guenther B, Manayani D, Mendy J, Smith J, Espinosa DA, Harris E, Alexander J, Vang L, Morello CS. Zika virus-like particle vaccine fusion loop mutation increases production yield but fails to protect AG129 mice against Zika virus challenge. PLoS Negl Trop Dis 2022; 16:e0010588. [PMID: 35793354 PMCID: PMC9292115 DOI: 10.1371/journal.pntd.0010588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/18/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus with maternal infection associated with preterm birth, congenital malformations, and fetal death, and adult infection associated with Guillain-Barré syndrome. Recent widespread endemic transmission of ZIKV and the potential for future outbreaks necessitate the development of an effective vaccine. We developed a ZIKV vaccine candidate based on virus-like-particles (VLPs) generated following transfection of mammalian HEK293T cells using a plasmid encoding the pre-membrane/membrane (prM/M) and envelope (E) structural protein genes. VLPs were collected from cell culture supernatant and purified by column chromatography with yields of approximately 1-2mg/L. To promote increased particle yields, a single amino acid change of phenylalanine to alanine was made in the E fusion loop at position 108 (F108A) of the lead VLP vaccine candidate. This mutation resulted in a modest 2-fold increase in F108A VLP production with no detectable prM processing by furin to a mature particle, in contrast to the lead candidate (parent). To evaluate immunogenicity and efficacy, AG129 mice were immunized with a dose titration of either the immature F108A or lead VLP (each alum adjuvanted). The resulting VLP-specific binding antibody (Ab) levels were comparable. However, geometric mean neutralizing Ab (nAb) titers using a recombinant ZIKV reporter were significantly lower with F108A immunization compared to lead. After virus challenge, all lead VLP-immunized groups showed a significant 3- to 4-Log10 reduction in mean ZIKV RNAemia levels compared with control mice immunized only with alum, but the RNAemia reduction of 0.5 Log10 for F108A groups was statistically similar to the control. Successful viral control by the lead VLP candidate following challenge supports further vaccine development for this candidate. Notably, nAb titer levels in the lead, but not F108A, VLP-immunized mice inversely correlated with RNAemia. Further evaluation of sera by an in vitro Ab-dependent enhancement assay demonstrated that the F108A VLP-induced immune sera had a significantly higher capacity to promote ZIKV infection in FcγR-expressing cells. These data indicate that a single amino acid change in the fusion loop resulted in increased VLP yields but that the immature F108A particles were significantly diminished in their capacity to induce nAbs and provide protection against ZIKV challenge. Zika virus (ZIKV) is transmitted by mosquitoes and is a serious health threat due to potential epidemic spread. Infection in adults may lead to Guillain-Barré syndrome, a neurological disorder, or may cause harm to a developing fetus resulting in preterm birth, fetal death, or devastating congenital malformations. There are currently no approved vaccines against ZIKV. We previously developed a lead candidate vaccine based on a virus-like particle (VLP) that was generated in tissue culture. This ZIKV shell is devoid of any viral genetic material. In previous studies, this lead VLP candidate generated neutralizing antibodies (nAbs) that recognized wild-type ZIKV and prevented viral replication in both mice and non-human primates. To increase production of the lead VLP candidate and decrease cost-of-goods, we introduced a single amino acid change, phenylalanine to alanine, in the envelope glycoprotein. This change resulted in a modest increase in VLP yield. However, this single amino acid change resulted in reduced induction of nAbs following immunization and no significant reduction of RNAemia following challenge compared to the lead candidate. The results of this study suggest this investigational vaccine candidate is not suitable for further vaccine development and that ZIKV VLP maturation may have an important role in protection.
Collapse
Affiliation(s)
- Danielle Thompson
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Ben Guenther
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Darly Manayani
- PaxVax Inc., San Diego, California, United States of America
| | - Jason Mendy
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Jonathan Smith
- PaxVax Inc., San Diego, California, United States of America
| | - Diego A. Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Jeff Alexander
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
- PaxVax Inc., San Diego, California, United States of America
| | - Lo Vang
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | | |
Collapse
|
7
|
Damm D, Kostka K, Weingärtner C, Wagner JT, Rojas-Sánchez L, Gensberger-Reigl S, Sokolova V, Überla K, Epple M, Temchura V. Covalent coupling of HIV-1 glycoprotein trimers to biodegradable calcium phosphate nanoparticles via genetically encoded aldehyde-tags. Acta Biomater 2022; 140:586-600. [PMID: 34968725 DOI: 10.1016/j.actbio.2021.12.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/14/2021] [Accepted: 12/20/2021] [Indexed: 01/02/2023]
Abstract
The usage of antigen-functionalized nanoparticles has become a major focus in the field of experimental HIV-1 vaccine research during the last decade. Various molecular mechanisms to couple native-like trimers of the HIV-1 envelope protein (Env) onto nanoparticle surfaces have been reported, but many come with disadvantages regarding the coupling efficiency and stability. In this study, a short amino acid sequence ("aldehyde-tag") was introduced at the C-terminus of a conformationally stabilized native-like Env. The post-translational conversion of a tag-associated cysteine to formylglycine creates a site-specific aldehyde group without alteration of the Env antigenicity. This aldehyde group was further utilized for bioconjugation of Env trimers. We demonstrated that the low acidic environment necessary for this bioconjugation is not affecting the trimer conformation. Furthermore, we developed a two-step coupling method for pH-sensitive nanoparticles. To this end, we conjugated aldehyde-tagged Env with Propargyl-PEG3-aminooxy linker (oxime ligation; Step-one) and coupled these conjugates by copper-catalyzed azide-alkyne cycloaddition (Click reaction; Step-two) to calcium phosphate nanoparticles (CaPs) functionalized with terminal azide groups. CaPs displaying orthogonally arranged Env trimers on their surface (o-CaPs) were superior in activation of Env-specific B-cells (in vitro) and induction of Env-specific antibody responses (in vivo) compared to CaPs with Env trimers coupled in a randomly oriented manner. Taken together, we present a reliable method for the site-specific, covalent coupling of HIV-1 Env native-like trimers to the surface of nanoparticle delivery systems. This method can be broadly applied for functionalization of nanoparticle platforms with conformationally stabilized candidate antigens for both vaccination and diagnostic approaches. STATEMENT OF SIGNIFICANCE: During the last decade antigen-functionalized nanoparticles have become a major focus in the field of experimental HIV-1 vaccines. Rational design led to the production of conformationally stabilized HIV-1 envelope protein (Env) trimers - the only target for the humoral immune system. Various molecular mechanisms to couple Env trimers onto nanoparticle surfaces have been reported, but many come with disadvantages regarding the coupling efficiency and stability. In this paper, we describe a highly selective bio-conjugation of Env trimers to the surface of medically relevant calcium phosphate nanoparticles. This method maintains the native-like protein conformation and has a broad potential application in functionalization of nanoparticle platforms with stabilized candidate antigens (including stabilized spike proteins of coronaviruses) for both vaccination and diagnostic approaches.
Collapse
Affiliation(s)
- D Damm
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - K Kostka
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstraße 5-7, 45141 Essen, Germany
| | - C Weingärtner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - J T Wagner
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - L Rojas-Sánchez
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstraße 5-7, 45141 Essen, Germany
| | - S Gensberger-Reigl
- Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nuremberg, Nikolaus-Fiebiger-Str. 10, 91058 Erlangen, Germany
| | - V Sokolova
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstraße 5-7, 45141 Essen, Germany
| | - K Überla
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - M Epple
- Inorganic Chemistry and Center for Nanointegration Duisburg-Essen (CeNIDE), University of Duisburg-Essen, Universitätsstraße 5-7, 45141 Essen, Germany
| | - V Temchura
- Institute of Clinical and Molecular Virology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Schlossgarten 4, 91054 Erlangen, Germany.
| |
Collapse
|
8
|
Kudlacek ST, Metz S, Thiono D, Payne AM, Phan TTN, Tian S, Forsberg LJ, Maguire J, Seim I, Zhang S, Tripathy A, Harrison J, Nicely NI, Soman S, McCracken MK, Gromowski GD, Jarman RG, Premkumar L, de Silva AM, Kuhlman B. Designed, highly expressing, thermostable dengue virus 2 envelope protein dimers elicit quaternary epitope antibodies. SCIENCE ADVANCES 2021; 7:eabg4084. [PMID: 34652943 PMCID: PMC8519570 DOI: 10.1126/sciadv.abg4084] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 08/25/2021] [Indexed: 05/30/2023]
Abstract
Dengue virus (DENV) is a worldwide health burden, and a safe vaccine is needed. Neutralizing antibodies bind to quaternary epitopes on DENV envelope (E) protein homodimers. However, recombinantly expressed soluble E proteins are monomers under vaccination conditions and do not present these quaternary epitopes, partly explaining their limited success as vaccine antigens. Using molecular modeling, we found DENV2 E protein mutations that induce dimerization at low concentrations (<100 pM) and enhance production yield by more than 50-fold. Cross-dimer epitope antibodies bind to the stabilized dimers, and a crystal structure resembles the wild-type (WT) E protein bound to a dimer epitope antibody. Mice immunized with the stabilized dimers developed antibodies that bind to E dimers and not monomers and elicited higher levels of DENV2-neutralizing antibodies compared to mice immunized with WT E antigen. Our findings demonstrate the feasibility of using structure-based design to produce subunit vaccines for dengue and other flaviviruses.
Collapse
Affiliation(s)
- Stephan T. Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Stefan Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Devina Thiono
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Alexander M. Payne
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Thanh T. N. Phan
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Lawrence J. Forsberg
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Jack Maguire
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ian Seim
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Shu Zhang
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ashutosh Tripathy
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Joseph Harrison
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Nathan I. Nicely
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Sandrine Soman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Michael K. McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Gregory D. Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Richard G. Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27514, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27514, USA
| |
Collapse
|
9
|
A Novel Antigenic Site Spanning Domains I and III of the Zika Virus Envelope Glycoprotein Is the Target of Strongly Neutralizing Human Monoclonal Antibodies. J Virol 2021; 95:JVI.02423-20. [PMID: 33597214 PMCID: PMC8104094 DOI: 10.1128/jvi.02423-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/05/2021] [Indexed: 12/18/2022] Open
Abstract
People infected with Zika virus develop durable neutralizing antibodies that prevent repeat infections. In the current study, we characterize a ZIKV-neutralizing human monoclonal antibody isolated from a patient after recovery. Our studies establish a novel site on the viral envelope that is targeted by human neutralizing antibodies. Our results are relevant to understanding how antibodies block infection and to guiding the design and evaluation of candidate vaccines. Zika virus (ZIKV), a mosquito-transmitted flavivirus, caused a large epidemic in Latin America between 2015 and 2017. Effective ZIKV vaccines and treatments are urgently needed to prevent future epidemics and severe disease sequelae. People infected with ZIKV develop strongly neutralizing antibodies linked to viral clearance and durable protective immunity. To understand the mechanisms of protective immunity and to support the development of ZIKV vaccines, we characterize here a strongly neutralizing antibody, B11F, isolated from a patient who recovered from ZIKV. Our results indicate that B11F targets a complex epitope on the virus that spans domains I and III of the envelope glycoprotein. While previous studies point to quaternary epitopes centered on domain II of the ZIKV E glycoprotein as targets of strongly neutralizing and protective human antibodies, we uncover a new site spanning domains I and III as a target of strongly neutralizing human antibodies. IMPORTANCE People infected with Zika virus develop durable neutralizing antibodies that prevent repeat infections. In the current study, we characterize a ZIKV-neutralizing human monoclonal antibody isolated from a patient after recovery. Our studies establish a novel site on the viral envelope that is targeted by human neutralizing antibodies. Our results are relevant to understanding how antibodies block infection and to guiding the design and evaluation of candidate vaccines.
Collapse
|
10
|
Dussupt V, Modjarrad K, Krebs SJ. Landscape of Monoclonal Antibodies Targeting Zika and Dengue: Therapeutic Solutions and Critical Insights for Vaccine Development. Front Immunol 2021; 11:621043. [PMID: 33664734 PMCID: PMC7921836 DOI: 10.3389/fimmu.2020.621043] [Citation(s) in RCA: 14] [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: 10/24/2020] [Accepted: 12/14/2020] [Indexed: 01/23/2023] Open
Abstract
The unprecedented 2015-2016 Zika outbreak in the Americas sparked global concern and drove the rapid deployment of vaccine and therapeutic countermeasures against this re-emerging pathogen. Alongside vaccine development, a number of potent neutralizing antibodies against Zika and related flaviviruses have been identified in recent years. High-throughput antibody isolation approaches have contributed to a better understanding of the B cell responses elicited following infection and/or vaccination. Structure-based approaches have illuminated species-specific and cross-protective epitopes of therapeutic value. This review will highlight previously described monoclonal antibodies with the best therapeutic potential against ZIKV and related flaviviruses, and discuss their implications for the rational design of better vaccine strategies.
Collapse
Affiliation(s)
- Vincent Dussupt
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Shelly J. Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| |
Collapse
|
11
|
Young E, Carnahan RH, Andrade DV, Kose N, Nargi RS, Fritch EJ, Munt JE, Doyle MP, White L, Baric TJ, Stoops M, DeSilva A, Tse LV, Martinez DR, Zhu D, Metz S, Wong MP, Espinosa DA, Montoya M, Biering SB, Sukulpolvi-Petty S, Kuan G, Balmaseda A, Diamond MS, Harris E, Crowe JE, Baric RS. Identification of Dengue Virus Serotype 3 Specific Antigenic Sites Targeted by Neutralizing Human Antibodies. Cell Host Microbe 2021; 27:710-724.e7. [PMID: 32407709 PMCID: PMC7309352 DOI: 10.1016/j.chom.2020.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/18/2020] [Accepted: 04/06/2020] [Indexed: 12/31/2022]
Abstract
The rational design of dengue virus (DENV) vaccines requires a detailed understanding of the molecular basis for antibody-mediated immunity. The durably protective antibody response to DENV after primary infection is serotype specific. However, there is an incomplete understanding of the antigenic determinants for DENV type-specific (TS) antibodies, especially for DENV serotype 3, which has only one well-studied, strongly neutralizing human monoclonal antibody (mAb). Here, we investigated the human B cell response in children after natural DENV infection in the endemic area of Nicaragua and isolated 15 DENV3 TS mAbs recognizing the envelope (E) glycoprotein. Functional epitope mapping of these mAbs and small animal prophylaxis studies revealed a complex landscape with protective epitopes clustering in at least 6-7 antigenic sites. Potently neutralizing TS mAbs recognized sites principally in E glycoprotein domains I and II, and patterns suggest frequent recognition of quaternary structures on the surface of viral particles.
Collapse
Affiliation(s)
- Ellen Young
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Robert H Carnahan
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Daniela V Andrade
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel S Nargi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan J Fritch
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Michael P Doyle
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura White
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Thomas J Baric
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Mark Stoops
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Aravinda DeSilva
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Longping V Tse
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - David R Martinez
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Deanna Zhu
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Stefan Metz
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Diego A Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Magelda Montoya
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Soila Sukulpolvi-Petty
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Guillermina Kuan
- Health Center Sócrates Flores Vivas, Ministry of Health, Managua, Nicaragua
| | - Angel Balmaseda
- National Virology Laboratory, National Center for Diagnosis and Reference, Ministry of Health, Managua, Nicaragua
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA.
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Ralph S Baric
- Department of Epidemiology, Gillings School of Public Health, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
12
|
Dengue Vaccines: The Promise and Pitfalls of Antibody-Mediated Protection. Cell Host Microbe 2021; 29:13-22. [PMID: 33444553 DOI: 10.1016/j.chom.2020.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/12/2020] [Accepted: 12/10/2020] [Indexed: 01/16/2023]
Abstract
More than 390 million human dengue virus (DENV) infections occur each year, worldwide. Dengvaxia, a live-virus tetravalent vaccine from Sanofi Pasteur, was recently approved for human clinical use, although vaccine performance against the four DENV serotypes is highly variable. Other dengue vaccines in advanced clinical testing also demonstrate variability in efficacy. In this review, we outline the benefits and challenges of developing a safe, effective, and balanced DENV vaccine that can provide uniform protection against all four serotypes. Even though T cell biology plays an important role in establishing protective immunity, this review focuses on B cell responses. We discuss the leading dengue vaccine candidates and review the specificity of antibody responses and the known immune correlates of protection against DENV infection. A better understanding of immune correlates of protection against DENV infection will inform the development of a vaccine that can provide long-term, uniform protection.
Collapse
|
13
|
Hobson-Peters J, Harrison JJ, Watterson D, Hazlewood JE, Vet LJ, Newton ND, Warrilow D, Colmant AMG, Taylor C, Huang B, Piyasena TBH, Chow WK, Setoh YX, Tang B, Nakayama E, Yan K, Amarilla AA, Wheatley S, Moore PR, Finger M, Kurucz N, Modhiran N, Young PR, Khromykh AA, Bielefeldt-Ohmann H, Suhrbier A, Hall RA. A recombinant platform for flavivirus vaccines and diagnostics using chimeras of a new insect-specific virus. Sci Transl Med 2020; 11:11/522/eaax7888. [PMID: 31826984 DOI: 10.1126/scitranslmed.aax7888] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/11/2019] [Indexed: 12/15/2022]
Abstract
Flaviviruses such as dengue, yellow fever, Zika, West Nile, and Japanese encephalitis virus present substantial global health burdens. New vaccines are being sought to address safety and manufacturing issues associated with current live attenuated vaccines. Here, we describe a new insect-specific flavivirus, Binjari virus, which was found to be remarkably tolerant for exchange of its structural protein genes (prME) with those of the aforementioned pathogenic vertebrate-infecting flaviviruses (VIFs). Chimeric BinJ/VIF-prME viruses remained replication defective in vertebrate cells but replicated with high efficiency in mosquito cells. Cryo-electron microscopy and monoclonal antibody binding studies illustrated that the chimeric BinJ/VIF-prME virus particles were structurally and immunologically similar to their parental VIFs. Pilot manufacturing in C6/36 cells suggests that high yields can be reached up to 109.5 cell culture infectious dose/ml or ≈7 mg/liter. BinJ/VIF-prME viruses showed utility in diagnostic (microsphere immunoassays and ELISAs using panels of human and equine sera) and vaccine applications (illustrating protection against Zika virus challenge in murine IFNAR-/- mouse models). BinJ/VIF-prME viruses thus represent a versatile, noninfectious (for vertebrate cells), high-yield technology for generating chimeric flavivirus particles with low biocontainment requirements.
Collapse
Affiliation(s)
- Jody Hobson-Peters
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia.
| | - Jessica J Harrison
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Daniel Watterson
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Jessamine E Hazlewood
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Laura J Vet
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Natalee D Newton
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - David Warrilow
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Agathe M G Colmant
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Carmel Taylor
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Bixing Huang
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Thisun B H Piyasena
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Weng Kong Chow
- Australian Defence Force Malaria and Infectious Disease Institute, Gallipoli Barracks, Queensland, Australia
| | - Yin Xiang Setoh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Bing Tang
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Eri Nakayama
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kexin Yan
- Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Alberto A Amarilla
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Sarah Wheatley
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Peter R Moore
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Mitchell Finger
- Public Health Virology Laboratory, Department of Health, Queensland Government, PO Box 594, Archerfield, Queensland, Australia
| | - Nina Kurucz
- Centre for Disease Control, Health Protection Division, Northern Territory Department of Health, Darwin, Northern Territory, Australia
| | - Naphak Modhiran
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Paul R Young
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Alexander A Khromykh
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia
| | - Helle Bielefeldt-Ohmann
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia.,School of Veterinary Science, University of Queensland Gatton Campus, Queensland 4343, Australia
| | - Andreas Suhrbier
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia.,Inflammation Biology Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland 4029, Australia
| | - Roy A Hall
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland, Australia.
| |
Collapse
|
14
|
Thomas A, Thiono DJ, Kudlacek ST, Forsberg J, Premkumar L, Tian S, Kuhlman B, de Silva AM, Metz SW. Dimerization of Dengue Virus E Subunits Impacts Antibody Function and Domain Focus. J Virol 2020; 94:e00745-20. [PMID: 32611757 PMCID: PMC7459570 DOI: 10.1128/jvi.00745-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Dengue virus (DENV) is responsible for the most prevalent and significant arthropod-borne viral infection of humans. The leading DENV vaccines are based on tetravalent live-attenuated virus platforms. In practice, it has been challenging to induce balanced and effective responses to each of the four DENV serotypes because of differences in the replication efficiency and immunogenicity of individual vaccine components. Unlike live vaccines, tetravalent DENV envelope (E) protein subunit vaccines are likely to stimulate balanced immune responses, because immunogenicity is replication independent. However, E protein subunit vaccines have historically performed poorly, in part because the antigens utilized were mainly monomers that did not display quaternary-structure epitopes found on E dimers and higher-order structures that form the viral envelope. In this study, we compared the immunogenicity of DENV2 E homodimers and DENV2 E monomers. The stabilized DENV2 homodimers, but not monomers, were efficiently recognized by virus-specific and flavivirus cross-reactive potently neutralizing antibodies that have been mapped to quaternary-structure epitopes displayed on the viral surface. In mice, the dimers stimulated 3-fold-higher levels of virus-specific neutralizing IgG that recognized epitopes different from those recognized by lower-level neutralizing antibodies induced by monomers. The dimer induced a stronger E domain I (EDI)- and EDII-targeted response, while the monomer antigens stimulated an EDIII epitope response and induced fusion loop epitope antibodies that are known to facilitate antibody-dependent enhancement (ADE). This study shows that DENV E subunit antigens that have been designed to mimic the structural organization of the viral surface are better vaccine antigens than E protein monomers.IMPORTANCE Dengue virus vaccine development is particularly challenging because vaccines have to provide protection against four different dengue virus stereotypes. The leading dengue virus vaccine candidates in clinical testing are all based on live-virus vaccine platforms and struggle to induce balanced immunity. Envelope subunit antigens have the potential to overcome these limitations but have historically performed poorly as vaccine antigens, because the versions tested previously were presented as monomers and not in their natural dimer configuration. This study shows that the authentic presentation of DENV2 E-based subunits has a strong impact on antibody responses, underscoring the importance of mimicking the complex protein structures that are found on DENV particle surfaces when designing subunit vaccines.
Collapse
Affiliation(s)
- Ashlie Thomas
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Devina J Thiono
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Stephan T Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - John Forsberg
- Department of Pharmacology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Brian Kuhlman
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Stefan W Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
15
|
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: 1.0] [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
|
16
|
Hosseini S, Muñoz-Soto RB, Oliva-Ramírez J, Vázquez-Villegas P, Aghamohammadi N, Rodriguez-Garcia A, Martinez-Chapa SO. Latest Updates in Dengue Fever Therapeutics: Natural, Marine and Synthetic Drugs. Curr Med Chem 2020; 27:719-744. [DOI: 10.2174/0929867325666180629124709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/25/2018] [Accepted: 06/01/2018] [Indexed: 11/22/2022]
Abstract
In this paper, we review the history of Dengue, the mechanism of infection, the
molecular characteristics and components of Dengue, the mechanism of entry to the target
cells, cyclization of the genome and replication process, as well as translation of the proteins
for virus assembly. The major emphasis of this work is on natural products and plant extracts,
which were used for as palliative or adjuvant treatment of Dengue. This review article also
summarizes the latest findings in regards to the marine products as effective drugs to target
different symptoms of Dengue. Furthermore, an update on synthetic drugs for treating Dengue
is provided in this review. As a novel alternative, we describe monoclonal antibody therapy
for Dengue management and treatment.
Collapse
Affiliation(s)
- Samira Hosseini
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, N.L. 64849, Mexico
| | - Rodrigo B. Muñoz-Soto
- Tecnologico de Monterrey, Campus Ciudad de México, Escuela de Ingeniería y Ciencias, Calle del Puente 222, Mexico City, Mexico
| | - Jacqueline Oliva-Ramírez
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Ave. Lago de Guadalupe Km 3.5, Cd Lopez Mateos, Atizapan, Estado de Mexico, Mexico
| | | | - Nasrin Aghamohammadi
- Centre for Occupational and Environmental Health, Department of Social and Preventive Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Aida Rodriguez-Garcia
- Universidad Autonoma de Nuevo Leon, Facultad de Ciencias Biologicas, Instituto de Biotecnología. Ave. Pedro de Alba S/N, Ciudad Universitaria, San Nicolás de los Garza, N.L. 66455, Mexico
| | | |
Collapse
|
17
|
Wilder-Smith A, Preet R, Brickley EB, Ximenes RADA, Miranda-Filho DDB, Turchi Martelli CM, Araújo TVBD, Montarroyos UR, Moreira ME, Turchi MD, Solomon T, Jacobs BC, Villamizar CP, Osorio L, de Filipps AMB, Neyts J, Kaptein S, Huits R, Ariën KK, Willison HJ, Edgar JM, Barnett SC, Peeling R, Boeras D, Guzman MG, de Silva AM, Falconar AK, Romero-Vivas C, Gaunt MW, Sette A, Weiskopf D, Lambrechts L, Dolk H, Morris JK, Orioli IM, O'Reilly KM, Yakob L, Rocklöv J, Soares C, Ferreira MLB, Franca RFDO, Precioso AR, Logan J, Lang T, Jamieson N, Massad E. ZikaPLAN: addressing the knowledge gaps and working towards a research preparedness network in the Americas. Glob Health Action 2020; 12:1666566. [PMID: 31640505 PMCID: PMC6818126 DOI: 10.1080/16549716.2019.1666566] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Zika Preparedness Latin American Network (ZikaPLAN) is a research consortium funded by the European Commission to address the research gaps in combating Zika and to establish a sustainable network with research capacity building in the Americas. Here we present a report on ZikaPLAN`s mid-term achievements since its initiation in October 2016 to June 2019, illustrating the research objectives of the 15 work packages ranging from virology, diagnostics, entomology and vector control, modelling to clinical cohort studies in pregnant women and neonates, as well as studies on the neurological complications of Zika infections in adolescents and adults. For example, the Neuroviruses Emerging in the Americas Study (NEAS) has set up more than 10 clinical sites in Colombia. Through the Butantan Phase 3 dengue vaccine trial, we have access to samples of 17,000 subjects in 14 different geographic locations in Brazil. To address the lack of access to clinical samples for diagnostic evaluation, ZikaPLAN set up a network of quality sites with access to well-characterized clinical specimens and capacity for independent evaluations. The International Committee for Congenital Anomaly Surveillance Tools was formed with global representation from regional networks conducting birth defects surveillance. We have collated a comprehensive inventory of resources and tools for birth defects surveillance, and developed an App for low resource regions facilitating the coding and description of all major externally visible congenital anomalies including congenital Zika syndrome. Research Capacity Network (REDe) is a shared and open resource centre where researchers and health workers can access tools, resources and support, enabling better and more research in the region. Addressing the gap in research capacity in LMICs is pivotal in ensuring broad-based systems to be prepared for the next outbreak. Our shared and open research space through REDe will be used to maximize the transfer of research into practice by summarizing the research output and by hosting the tools, resources, guidance and recommendations generated by these studies. Leveraging on the research from this consortium, we are working towards a research preparedness network.
Collapse
Affiliation(s)
| | - Raman Preet
- Department of Epidemiology and Global Health, Umeå University , Umeå , Sweden
| | | | - Ricardo Arraes de Alencar Ximenes
- Departamento de Medicina Tropical, Universidade Federal de Pernambuco , Recife , Brasil.,Departamento de Medicina Interna, Universidade de Pernambuco , Recife , Brasil
| | | | | | | | | | | | - Marília Dalva Turchi
- Instituto de Patologia Tropical e Saúde Publica, Universidade Federal de Goiás , Goiânia , Brasil
| | - Tom Solomon
- Institute of Infection and Global Health, The University of Liverpool , Liverpool , UK
| | - Bart C Jacobs
- Departments of Neurology and Immunology, Erasmus Universitair Medisch Centrum Rotterdam , The Netherlands
| | | | | | | | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute of Medical Research , Leuven , Belgium
| | - Suzanne Kaptein
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Rega Institute of Medical Research , Leuven , Belgium
| | - Ralph Huits
- Institute of Tropical Medicine , Antwerp , Belgium
| | | | - Hugh J Willison
- Institute of Infection, Immunity & Inflammation, University of Glasgow , Glasgow , UK
| | - Julia M Edgar
- Institute of Infection, Immunity & Inflammation, University of Glasgow , Glasgow , UK
| | - Susan C Barnett
- Institute of Infection, Immunity & Inflammation, University of Glasgow , Glasgow , UK
| | | | - Debi Boeras
- London School of Hygiene & Tropical Medicine , London , UK
| | | | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill , NC , USA
| | - Andrew K Falconar
- London School of Hygiene & Tropical Medicine , London , UK.,Departmento del Medicina, Fundacion Universidad del Norte , Barranquilla , Colombia
| | - Claudia Romero-Vivas
- Departmento del Medicina, Fundacion Universidad del Norte , Barranquilla , Colombia
| | | | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology , La Jolla , CA , USA.,Department of Medicine, University of California San Diego , La Jolla , CA , USA
| | - Daniela Weiskopf
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology , La Jolla , CA , USA
| | - Louis Lambrechts
- Insect-Virus Interactions Unit, Institut Pasteur, UMR2000, CNRS , Paris , France
| | - Helen Dolk
- Maternal Fetal and Infant Research Centre, Institute of Nursing and Health Research, Ulster University , Newtownabbey , UK
| | - Joan K Morris
- Population Health Research Institute, St George's, University of London , London , UK
| | - Ieda M Orioli
- Associação Técnico-Científica Estudo Colaborativo Latino Americano de Malformações Congênitas (ECLAMC) no Departmento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro , Rio de Janeiro , Brazil
| | | | - Laith Yakob
- London School of Hygiene & Tropical Medicine , London , UK
| | - Joacim Rocklöv
- Department of Epidemiology and Global Health, Umeå University , Umeå , Sweden
| | - Cristiane Soares
- Hospital Federal dos Servidores do Estado , Rio de Janeiro , Brazil
| | | | | | - Alexander R Precioso
- Instituto Butantan , Brazil.,Pediatrics Department, Medical School of University of Sao Paulo , Sao Paulo , Brazil
| | - James Logan
- London School of Hygiene & Tropical Medicine , London , UK
| | - Trudie Lang
- The Global Health Network, Masters and Scholars of the University of Oxford , Oxford , UK
| | - Nina Jamieson
- The Global Health Network, Masters and Scholars of the University of Oxford , Oxford , UK
| | - Eduardo Massad
- Fundacao de Apoio a Universidade de Sao Paulo , Sao Paulo , Brazil.,School of Applied Mathematics, Fundacao Getulio Vargas , Rio de Janeiro , Brazil
| |
Collapse
|
18
|
Metz SW, Thomas A, Brackbill A, Forsberg J, Miley MJ, Lopez CA, Lazear HM, Tian S, de Silva AM. Oligomeric state of the ZIKV E protein defines protective immune responses. Nat Commun 2019; 10:4606. [PMID: 31601808 PMCID: PMC6787251 DOI: 10.1038/s41467-019-12677-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
The current leading Zika vaccine candidates in clinical testing are based on live or killed virus platforms, which have safety issues, especially in pregnant women. Zika subunit vaccines, however, have shown poor performance in preclinical studies, most likely because the antigens tested do not display critical quaternary structure epitopes present on Zika E protein homodimers that cover the surface of the virus. Here, we produce stable recombinant E protein homodimers that are recognized by strongly neutralizing Zika specific monoclonal antibodies. In mice, the dimeric antigen stimulate strongly neutralizing antibodies that target epitopes that are similar to epitopes recognized by human antibodies following natural Zika virus infection. The monomer antigen stimulates low levels of E-domain III targeting neutralizing antibodies. In a Zika challenge model, only E dimer antigen stimulates protective antibodies, not the monomer. These results highlight the importance of mimicking the highly structured flavivirus surface when designing subunit vaccines.
Collapse
Affiliation(s)
- Stefan W Metz
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Ashlie Thomas
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alex Brackbill
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John Forsberg
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael J Miley
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Cesar A Lopez
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Helen M Lazear
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
19
|
Kudlacek ST, Metz SW. Focused dengue vaccine development: outwitting nature's design. Pathog Dis 2019; 77:5307883. [PMID: 30726906 DOI: 10.1093/femspd/ftz003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/15/2019] [Indexed: 12/28/2022] Open
Abstract
The four DENV serotypes are mosquito-borne pathogens that belong to the Flavivirus genus. These viruses present a major global health burden, being endemic in over 120 countries, causing ∼390 million reported infections yearly, with clinical symptoms ranging from mild fever to severe and potentially fatal hemorrhagic syndromes. Development of a safe and efficacious DENV vaccine is challenging because of the need to induce immunity against all four serotypes simultaneously, as immunity against one serotype can potentially enhance disease caused by a heterotypic secondary infection. So far, live-virus particle-based vaccine approaches struggle with inducing protective tetravalent immunity, while recombinant subunit approaches that use the envelope protein (E) as the major antigen, are gaining promise in preclinical studies. However, E-based subunits require further development and characterization to be used as effective vaccine antigens against DENV. In this review, we will address the shortcomings of recombinant E-based antigens and will discuss potential solutions to enhance E-based subunit antigen immunogenicity and vaccine efficacy.
Collapse
Affiliation(s)
- Stephan T Kudlacek
- Department of Biochemistry and Biophysics, University of North Carolina, 125 Mason Farm Road, 6230E Marisco Hall, Chapel Hill, NC 27599, USA
| | - Stefan W Metz
- Department of Microbiology and Immunology, University of North Carolina, 125 Mason Farm Road, 6230E Marisco Hall, Chapel Hill, NC 27599, USA
| |
Collapse
|
20
|
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: 7.8] [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
|
21
|
Yang C, Zeng F, Gao X, Zhao S, Li X, Liu S, Li N, Deng C, Zhang B, Gong R. Characterization of two engineered dimeric Zika virus envelope proteins as immunogens for neutralizing antibody selection and vaccine design. J Biol Chem 2019; 294:10638-10648. [PMID: 31138647 DOI: 10.1074/jbc.ra119.007443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 05/22/2019] [Indexed: 01/07/2023] Open
Abstract
The envelope protein of Zika virus (ZIKV) exists as a dimer on the mature viral surface and is an attractive antiviral target because it mediates viral entry. However, recombinant soluble wild-type ZIKV envelope (wtZE) might preferentially exist as monomer (monZE). Recently, it has been shown that the A264C substitution could promote formation of dimeric ZIKV envelope protein (ZEA264C), requiring further characterization of purified ZEA264C for its potential applications in vaccine development. We also noted that ZEA264C, connected by disulfide bond, might be different from the noncovalent native envelope dimer on the virion surface. Because the antibody Fc fragment exists as dimer and is widely used for fusion protein construction, here we fused wtZE to human immunoglobulin G1 (IgG1) Fc fragment (ZE-Fc) for noncovalent wtZE dimerization. Using a multistep purification procedure, we separated dimeric ZEA264C and ZE-Fc, revealing that they both exhibit typical β-sheet-rich secondary structures and stabilities similar to those of monZE. The binding activities of monZE, ZEA264C, and ZE-Fc to neutralizing antibodies targeting different epitopes indicated that ZEA264C and ZE-Fc could better mimic the native dimeric status, especially in terms of the formation of tertiary and quaternary epitopes. Both ZEA264C and ZE-Fc recognize a ZIKV-sensitive cell line as does monZE, indicating that the two constructs are still functional. Furthermore, a murine immunization assay disclose that ZEA264C and ZE-Fc elicit more neutralizing antibody responses than monZE does. These results suggest that the two immunogen candidates ZEA264C and ZE-Fc have potential utility for neutralizing antibody selection and vaccine design against ZIKV.
Collapse
Affiliation(s)
- Chunpeng Yang
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China, and
| | - Fang Zeng
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China, and
| | - Xinyu Gao
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China, and
| | - Shaojuan Zhao
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China, and
| | - Xuan Li
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Sheng Liu
- Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics & Molecular Imaging Key Laboratory, Systems Biology Theme, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Na Li
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China.,University of Chinese Academy of Sciences, Beijing 100049, China, and
| | - Chenglin Deng
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Bo Zhang
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Rui Gong
- From the CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China,
| |
Collapse
|
22
|
Collins MH, Tu HA, Gimblet-Ochieng C, Liou GJA, Jadi RS, Metz SW, Thomas A, McElvany BD, Davidson E, Doranz BJ, Reyes Y, Bowman NM, Becker-Dreps S, Bucardo F, Lazear HM, Diehl SA, de Silva AM. Human antibody response to Zika targets type-specific quaternary structure epitopes. JCI Insight 2019; 4:124588. [PMID: 30996133 DOI: 10.1172/jci.insight.124588] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 03/07/2019] [Indexed: 12/22/2022] Open
Abstract
The recent Zika virus (ZIKV) epidemic in the Americas has revealed rare but serious manifestations of infection. ZIKV has emerged in regions endemic for dengue virus (DENV), a closely related mosquito-borne flavivirus. Cross-reactive antibodies confound studies of ZIKV epidemiology and pathogenesis. The immune responses to ZIKV may be different in people, depending on their DENV immune status. Here, we focus on the human B cell and antibody response to ZIKV as a primary flavivirus infection to define the properties of neutralizing and protective antibodies generated in the absence of preexisting immunity to DENV. The plasma antibody and memory B cell response is highly ZIKV type-specific, and ZIKV-neutralizing antibodies mainly target quaternary structure epitopes on the viral envelope. To map viral epitopes targeted by protective antibodies, we isolated 2 type-specific monoclonal antibodies (mAbs) from a ZIKV case. Both mAbs were strongly neutralizing in vitro and protective in vivo. The mAbs recognize distinct epitopes centered on domains I and II of the envelope protein. We also demonstrate that the epitopes of these mAbs define antigenic regions commonly targeted by plasma antibodies in individuals from endemic and nonendemic regions who have recovered from ZIKV infections.
Collapse
Affiliation(s)
- Matthew H Collins
- Department of Medicine, Emory University, Atlanta, Georgia, USA, and Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Decatur, Georgia, USA.,Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Huy A Tu
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, Vermont, USA.,Vaccine Testing Center, Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Ciara Gimblet-Ochieng
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Guei-Jiun Alice Liou
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Ramesh S Jadi
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Stefan W Metz
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Ashlie Thomas
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Benjamin D McElvany
- Vaccine Testing Center, Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Edgar Davidson
- Integral Molecular, Inc., Philadelphia, Pennsylvania, USA
| | | | - Yaoska Reyes
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of León, Nicaragua
| | - Natalie M Bowman
- Department of Medicine, Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Sylvia Becker-Dreps
- Departments of Family Medicine and Epidemiology, University of North Carolina at Chapel Hill, Schools of Medicine and Public Health, Chapel Hill, North Carolina, USA
| | - Filemón Bucardo
- Department of Microbiology, Faculty of Medical Sciences, National Autonomous University of León, Nicaragua
| | - Helen M Lazear
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Sean A Diehl
- Cellular, Molecular, and Biomedical Sciences Program, University of Vermont, Burlington, Vermont, USA.,Vaccine Testing Center, Department of Microbiology and Molecular Genetics, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Kim YC, Lopez-Camacho C, Nettleship JE, Rahman N, Hill ML, Silva-Reyes L, Ortiz-Martinez G, Figueroa-Aguilar G, Mar MA, Vivanco-Cid H, Rollier CS, Zitzmann N, Viveros-Sandoval ME, Owens RJ, Reyes-Sandoval A. Optimization of Zika virus envelope protein production for ELISA and correlation of antibody titers with virus neutralization in Mexican patients from an arbovirus endemic region. Virol J 2018; 15:193. [PMID: 30587198 PMCID: PMC6307127 DOI: 10.1186/s12985-018-1104-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 12/04/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Zika virus (ZIKV) has become a global threat with immediate need for accurate diagnostics, efficacious vaccines and therapeutics. Several ZIKV envelope (Env)-based vaccines have been developed recently. However, many commercially available ZIKV Env are based on the African lineage and produced in insect cells. Here, we sought to produce Asian-lineage ZIKV Env in mammalian cells for research and clinical applications. METHODS We designed various gene expression constructs to optimize the production of ZIKV using prM-Env and full or C-terminal truncations of Env; with or without a rat CD4 fusion partner to allow large-scale production of soluble protein in mammalian HEK293 cells. Protein expression was verified by mass spectrometry and western-blot with a pan-flavivirus antibody, a ZIKV Env monoclonal antibody and with immune sera from adenoviral (ChAdOx1) ZIKV Env-vaccinated mice. The resulting Env-CD4 was used as a coating reagent for immunoassay (ELISA) using both mouse and human seropositive sera. RESULTS Replacement of the C-terminus transmembrane Env domain by a rat CD4 and addition of prM supported optimal expression and secretion of Env. Binding between the antigens and the antibodies was similar to binding when using commercially available ZIKV Env reagents. Furthermore, antibodies from ZIKV patients bound ZIKV Env-CD4 in ELISA assays, whereas sera from healthy blood donors yielded minimal OD background. The serological outcomes of this assay correlated also with ZIKV neutralisation capacity in vitro. CONCLUSIONS Results obtained from this study indicate the potential of the Asian-lineage Zika Env-CD4 and Env proteins in ELISA assays to monitor humoral immune responses in upcoming clinical trials as well as a sero-diagnostic tool in ZIKV infection.
Collapse
Affiliation(s)
- Young Chan Kim
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, University of Oxford, Old Road Campus Research Building. Roosevelt Drive, Oxford, OX3 7DQ, UK.,Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK
| | - Cesar Lopez-Camacho
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, University of Oxford, Old Road Campus Research Building. Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Joanne E Nettleship
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.,Rutherford Appleton Laboratory, OPPF-UK, Research Complex at Harwell, Oxford, UK
| | - Nahid Rahman
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.,Rutherford Appleton Laboratory, OPPF-UK, Research Complex at Harwell, Oxford, UK
| | - Michelle L Hill
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Laura Silva-Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Georgina Ortiz-Martinez
- Laboratorio de Hemostasia y Biología Vascular. División de Estudios de Posgrado. Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, UMSNH, Morelia, Mexico.,UMSNH-Oxford University of Oxford Clinical Research Laboratory (UMOCRL), Faculty of Biological and Medical Sciences "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Gloria Figueroa-Aguilar
- Laboratorio Estatal de Salud Pública, Secretaría de Salud de Michoacán, Morelia, Michoacán, Mexico
| | - María Antonieta Mar
- HGZMF No. 12 Lázaro Cárdenas Michoacán dirección av. Lázaro Cárdenas No. 154 Col. Centro Lázaro Cárdenas Michoacán, Veracruz, Mexico
| | - Héctor Vivanco-Cid
- Instituto de Investigaciones Médico-Biológicas, Universidad Veracruzana, Veracruz, Mexico
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Centre for Clinical Vaccinology and Tropical Medicine, Churchill Hospital, Oxford, UK
| | - Nicole Zitzmann
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Martha Eva Viveros-Sandoval
- Laboratorio de Hemostasia y Biología Vascular. División de Estudios de Posgrado. Facultad de Ciencias Médicas y Biológicas "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, UMSNH, Morelia, Mexico.,UMSNH-Oxford University of Oxford Clinical Research Laboratory (UMOCRL), Faculty of Biological and Medical Sciences "Dr. Ignacio Chávez", Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, Mexico
| | - Raymond J Owens
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, UK.,Rutherford Appleton Laboratory, OPPF-UK, Research Complex at Harwell, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, The Henry Wellcome Building for Molecular Physiology, University of Oxford, Old Road Campus Research Building. Roosevelt Drive, Oxford, OX3 7DQ, UK.
| |
Collapse
|
24
|
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
|
25
|
Potent Neutralizing Human Monoclonal Antibodies Preferentially Target Mature Dengue Virus Particles: Implication for Novel Strategy for Dengue Vaccine. J Virol 2018; 92:JVI.00556-18. [PMID: 30185598 PMCID: PMC6232466 DOI: 10.1128/jvi.00556-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/22/2018] [Indexed: 11/20/2022] Open
Abstract
The four serotypes of dengue virus (DENV) cause the most important mosquito-borne viral disease in humans. The envelope (E) protein is the major target of neutralizing antibodies and contains 3 domains (domain I [DI], DII, and DIII). Recent studies reported that human monoclonal antibodies (MAbs) recognizing DIII, the D1/DII hinge, the E-dimer epitope, or a quaternary epitope involving DI/DII/DIII are more potently neutralizing than those recognizing the fusion loop (FL) of DII. Due to inefficient cleavage of the premembrane protein, DENV suspensions consist of a mixture of mature, immature, and partially immature particles. We investigated the neutralization and binding of 22 human MAbs to DENV serotype 1 (DENV1) virions with differential maturation status. Compared with FL MAbs, DIII, DI/DII hinge, and E-dimer epitope MAbs showed higher maximum binding and avidity to mature particles relative to immature particles; this feature may contribute to the strong neutralizing potency of such MAbs. FL-specific MAbs required 57 to 87% occupancy on mature particles to achieve half-maximal neutralization (NT50), whereas the potently neutralizing MAbs achieved NT50 states at 20 to 38% occupancy. Analysis of the MAb repertoire and polyclonal sera from patients with primary DENV1 infection supports the immunodominance of cross-reactive anti-E antibodies over type-specific antibodies. After depletion with viral particles from a heterologous DENV serotype, the type-specific neutralizing antibodies remained and showed binding features shared by potent neutralizing MAbs. Taken together, these findings suggest that the use of homogeneous mature DENV particles as an immunogen may induce more potent neutralizing antibodies against DENV than the use of immature or mixed particles.IMPORTANCE With an estimated 390 million infections per year, the four serotypes of dengue virus (DENV) cause the most important mosquito-borne viral disease in humans. The dengue vaccine Dengvaxia was licensed; however, its low efficacy among dengue-naive individuals and increased risk of causing severe dengue in children highlight the need for a better understanding of the role of human antibodies in immunity against DENV. DENV suspensions contain mature, immature, and partially immature particles. We investigated the binding of 22 human monoclonal antibodies (MAbs) to the DENV envelope protein on particles with different maturation states. Potently neutralizing MAbs had higher relative maximum binding and avidity to mature particles than weakly neutralizing MAbs. This was supported by analysis of MAb repertoires and polyclonal sera from patients with primary DENV infection. Together, these findings suggest that mature particles may be the optimal form of presentation of the envelope protein to induce more potent neutralizing antibodies against DENV.
Collapse
|
26
|
Shen WF, Galula JU, Liu JH, Liao MY, Huang CH, Wang YC, Wu HC, Liang JJ, Lin YL, Whitney MT, Chang GJJ, Chen SR, Wu SR, Chao DY. Epitope resurfacing on dengue virus-like particle vaccine preparation to induce broad neutralizing antibody. eLife 2018; 7:38970. [PMID: 30334522 PMCID: PMC6234032 DOI: 10.7554/elife.38970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/18/2018] [Indexed: 12/25/2022] Open
Abstract
Dengue fever is caused by four different serotypes of dengue virus (DENV) which is the leading cause of worldwide arboviral diseases in humans. Virus-like particles (VLPs) containing flavivirus prM/E proteins have been demonstrated to be a potential vaccine candidate; however, the structure of dengue VLP is poorly understood. Herein VLP derived from DENV serotype-2 were engineered becoming highly matured (mD2VLP) and showed variable size distribution with diameter of ~31 nm forming the major population under cryo-electron microscopy examination. Furthermore, mD2VLP particles of 31 nm diameter possess a T = 1 icosahedral symmetry with a groove located within the E-protein dimers near the 2-fold vertices that exposed highly overlapping, cryptic neutralizing epitopes. Mice vaccinated with mD2VLP generated higher cross-reactive (CR) neutralization antibodies (NtAbs) and were fully protected against all 4 serotypes of DENV. Our results highlight the potential of ‘epitope-resurfaced’ mature-form D2VLPs in inducing quaternary structure-recognizing broad CR NtAbs to guide future dengue vaccine design.
Collapse
Affiliation(s)
- Wen-Fan Shen
- Microbial Genomics Ph.D. Program, National Chung Hsing University and Academia Sinica, Taichung City, Taiwan
| | - Jedhan Ucat Galula
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Jyung-Hurng Liu
- Institute of Genomics and Bioinformatics, College of Life Science, National Chung-Hsing University, Taichung City, Taiwan
| | - Mei-Ying Liao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Cheng-Hao Huang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Yu-Chun Wang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Matthew T Whitney
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States
| | - Gwong-Jen J Chang
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States
| | - Sheng-Ren Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Rung Wu
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Day-Yu Chao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| |
Collapse
|
27
|
Harsh S, Ozakman Y, Kitchen SM, Paquin-Proulx D, Nixon DF, Eleftherianos I. Dicer-2 Regulates Resistance and Maintains Homeostasis against Zika Virus Infection in Drosophila. THE JOURNAL OF IMMUNOLOGY 2018; 201:3058-3072. [PMID: 30305326 DOI: 10.4049/jimmunol.1800597] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022]
Abstract
Zika virus (ZIKV) outbreaks pose a massive public health threat in several countries. We have developed an in vivo model to investigate the host-ZIKV interaction in Drosophila We have found that a strain of ZIKV replicates in wild-type flies without reducing their survival ability. We have shown that ZIKV infection triggers RNA interference and that mutating Dicer-2 results in enhanced ZIKV load and increased susceptibility to ZIKV infection. Using a flavivirus-specific Ab, we have found that ZIKV is localized in the gut and fat body cells of the infected wild-type flies and results in their perturbed homeostasis. In addition, Dicer-2 mutants display severely reduced insulin activity, which could contribute toward the increased mortality of these flies. Our work establishes the suitability of Drosophila as the model system to study host-ZIKV dynamics, which is expected to greatly advance our understanding of the molecular and physiological processes that determine the outcome of this disease.
Collapse
Affiliation(s)
- Sneh Harsh
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Yaprak Ozakman
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| | - Shannon M Kitchen
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Dominic Paquin-Proulx
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Douglas F Nixon
- Department of Microbiology, Immunology, and Tropical Medicine, GW School of Medicine & Health Sciences, The George Washington University, Washington, DC 20052
| | - Ioannis Eleftherianos
- Department of Biological Sciences, The Institute for Biomedical Sciences, The George Washington University, Washington, DC 20052; and
| |
Collapse
|
28
|
Metz SW, Thomas A, Brackbill A, Xianwen Y, Stone M, Horvath K, Miley MJ, Luft C, DeSimone JM, Tian S, de Silva AM. Nanoparticle delivery of a tetravalent E protein subunit vaccine induces balanced, type-specific neutralizing antibodies to each dengue virus serotype. PLoS Negl Trop Dis 2018; 12:e0006793. [PMID: 30248097 PMCID: PMC6171938 DOI: 10.1371/journal.pntd.0006793] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 10/04/2018] [Accepted: 08/27/2018] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV) is the causative agent of dengue fever and dengue hemorrhagic shock syndrome. Dengue vaccine development is challenging because of the need to induce protection against four antigenically distinct DENV serotypes. Recent studies indicate that tetravalent DENV vaccines must induce balanced, serotype-specific neutralizing antibodies to achieve durable protective immunity against all 4 serotypes. With the leading live attenuated tetravalent DENV vaccines, it has been difficult to achieve balanced and type-specific responses to each serotype, most likely because of unbalanced replication of vaccine viral strains. Here we evaluate a tetravalent DENV protein subunit vaccine, based on recombinant envelope protein (rE) adsorbed to the surface of poly (lactic-co-glycolic acid) (PLGA) nanoparticles for immunogenicity in mice. In monovalent and tetravalent formulations, we show that particulate rE induced higher neutralizing antibody titers compared to the soluble rE antigen alone. Importantly, we show the trend that tetravalent rE adsorbed to nanoparticles stimulated a more balanced serotype specific antibody response to each DENV serotype compared to soluble antigens. Our results demonstrate that tetravalent DENV subunit vaccines displayed on nanoparticles have the potential to overcome unbalanced immunity observed for leading live-attenuated vaccine candidates.
Collapse
Affiliation(s)
- Stefan W. Metz
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Ashlie Thomas
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Alex Brackbill
- Department of Pharmacology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Yi Xianwen
- Lineberger Comprehensive Center, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Michele Stone
- Liquidia Technologies, Research Triangle Park, Durham, NC, United States of America
| | - Katie Horvath
- Liquidia Technologies, Research Triangle Park, Durham, NC, United States of America
| | - Michael J. Miley
- Department of Pharmacology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Chris Luft
- Liquidia Technologies, Research Triangle Park, Durham, NC, United States of America
| | - Joseph M. DeSimone
- Liquidia Technologies, Research Triangle Park, Durham, NC, United States of America
- Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
- Department of Chemistry, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
| | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
- * E-mail: (ST); (AMdS)
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, Chapel Hill, NC, United States of America
- * E-mail: (ST); (AMdS)
| |
Collapse
|
29
|
Kudlacek ST, Premkumar L, Metz SW, Tripathy A, Bobkov AA, Payne AM, Graham S, Brackbill JA, Miley MJ, de Silva AM, Kuhlman B. Physiological temperatures reduce dimerization of dengue and Zika virus recombinant envelope proteins. J Biol Chem 2018; 293:8922-8933. [PMID: 29678884 PMCID: PMC5995514 DOI: 10.1074/jbc.ra118.002658] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/13/2018] [Indexed: 01/01/2023] Open
Abstract
The spread of dengue (DENV) and Zika virus (ZIKV) is a major public health concern. The primary target of antibodies that neutralize DENV and ZIKV is the envelope (E) glycoprotein, and there is interest in using soluble recombinant E (sRecE) proteins as subunit vaccines. However, the most potent neutralizing antibodies against DENV and ZIKV recognize epitopes on the virion surface that span two or more E proteins. Therefore, to create effective DENV and ZIKV vaccines, presentation of these quaternary epitopes may be necessary. The sRecE proteins from DENV and ZIKV crystallize as native-like dimers, but studies in solution suggest that these dimers are marginally stable. To better understand the challenges associated with creating stable sRecE dimers, we characterized the thermostability of sRecE proteins from ZIKV and three DENV serotypes, DENV2-4. All four proteins irreversibly unfolded at moderate temperatures (46-53 °C). At 23 °C and low micromolar concentrations, DENV2 and ZIKV were primarily dimeric, and DENV3-4 were primarily monomeric, whereas at 37 °C, all four proteins were predominantly monomeric. We further show that the dissociation constant for DENV2 dimerization is very temperature-sensitive, ranging from <1 μm at 25 °C to 50 μm at 41 °C, due to a large exothermic enthalpy of binding of -79 kcal/mol. We also found that quaternary epitope antibody binding to DENV2-4 and ZIKV sRecE is reduced at 37 °C. Our observation of reduced sRecE dimerization at physiological temperature highlights the need for stabilizing the dimer as part of its development as a subunit vaccine.
Collapse
Affiliation(s)
- Stephan T Kudlacek
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Lakshmanane Premkumar
- the Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Stefan W Metz
- the Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Ashutosh Tripathy
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Andrey A Bobkov
- the Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037
| | - Alexander Matthew Payne
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Stephen Graham
- the Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - James A Brackbill
- the Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, and
| | - Michael J Miley
- the Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599, and
| | - Aravinda M de Silva
- the Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599
| | - Brian Kuhlman
- From the Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, North Carolina 27599,
- the Lineburger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
30
|
Kim DTH, Bao DT, Park H, Ngoc NM, Yeo SJ. Development of a novel peptide aptamer-based immunoassay to detect Zika virus in serum and urine. Am J Cancer Res 2018; 8:3629-3642. [PMID: 30026871 PMCID: PMC6037026 DOI: 10.7150/thno.25955] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV) has been identified as a cause of adverse outcomes of pregnancy, including microcephaly and other congenital diseases. Most people infected with ZIKV do not show any symptoms. Development of a method to discriminate dengue virus (DENV) and ZIKV infections has been challenging, and efficient assays for patient management are limited, attributable to high levels of cross-reactivity among co-circulating Flaviviruses. Thus, there is an urgent need for a specific high-throughput diagnostic assay to discriminate ZIKV infections from other Flavivirus infections. Methods: A novel epitope peptide of the ZIKV envelope protein was predicted using three immune epitope database analysis tools and then further modified. A molecular docking study was conducted using three-dimensional structures of the ZIKV envelope and peptide. Experimentally, interactions between the selected peptides and virus were assessed via a fluorescence-linked sandwich immunosorbent assay (FLISA), and performance of peptide-linked sandwich FLISA was evaluated in virus-spiked human serum and urine. Results: The Z_10.8 peptide (KRAVVSCAEA) was predicted to be a suitable detector, with a higher binding affinity than other candidates based on four criteria (binding affinity, root mean square deviation, position of amine residue of lysine at the N-terminus, and interactive site) in a docking study. Z_10.8 was significantly more efficient at detecting ZIKV than the other two peptides, as shown in the direct FLISA (P < 0.001). Further, the equilibrium dissociation constant (Kd) for the Z_10.8 peptide was 706.0 ± 177.9 (mean ± SD, nM), with specificity to discriminate ZIKV from DENV. The limit of detection for the sandwich FLISA was calculated as 1×104 tissue culture infective dose (TCID)50/mL. The presence of serum or urine did not interfere with the performance of the Z_10.8-linked sandwich FLISA. Conclusion: Four criteria are suggested for the development of an in silico modeled peptide aptamer; this computerized peptide aptamer discriminated ZIKV from DENV via immunoassay.
Collapse
|
31
|
Coffman JE, Metz SW, Brackbill A, Paul M, Miley MJ, DeSimone J, Luft JC, de Silva A, Tian S. Optimization of Surface Display of DENV2 E Protein on a Nanoparticle to Induce Virus Specific Neutralizing Antibody Responses. Bioconjug Chem 2018; 29:1544-1552. [DOI: 10.1021/acs.bioconjchem.8b00090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Jason E. Coffman
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27607, United States
| | | | | | | | | | - Joseph DeSimone
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27607, United States
| | | | | | | |
Collapse
|
32
|
Gallichotte EN, Baric TJ, Yount BL, Widman DG, Durbin A, Whitehead S, Baric RS, de Silva AM. Human dengue virus serotype 2 neutralizing antibodies target two distinct quaternary epitopes. PLoS Pathog 2018; 14:e1006934. [PMID: 29481552 PMCID: PMC5843351 DOI: 10.1371/journal.ppat.1006934] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 03/08/2018] [Accepted: 02/12/2018] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV) infection causes dengue fever, dengue hemorrhagic fever and dengue shock syndrome. It is estimated that a third of the world’s population is at risk for infection, with an estimated 390 million infections annually. Dengue virus serotype 2 (DENV2) causes severe epidemics, and the leading tetravalent dengue vaccine has lower efficacy against DENV2 compared to the other 3 serotypes. In natural DENV2 infections, strongly neutralizing type-specific antibodies provide protection against subsequent DENV2 infection. While the epitopes of some human DENV2 type-specific antibodies have been mapped, it is not known if these are representative of the polyclonal antibody response. Using structure-guided immunogen design and reverse genetics, we generated a panel of recombinant viruses containing amino acid alterations and epitope transplants between different serotypes. Using this panel of recombinant viruses in binding, competition, and neutralization assays, we have finely mapped the epitopes of three human DENV2 type-specific monoclonal antibodies, finding shared and distinct epitope regions. Additionally, we used these recombinant viruses and polyclonal sera to dissect the epitope-specific responses following primary DENV2 natural infection and monovalent vaccination. Our results demonstrate that antibodies raised following DENV2 infection or vaccination circulate as separate populations that neutralize by occupying domain III and domain I quaternary epitopes. The fraction of neutralizing antibodies directed to different epitopes differs between individuals. The identification of these epitopes could potentially be harnessed to evaluate epitope-specific antibody responses as correlates of protective immunity, potentially improving vaccine design. Dengue viruses (DENV) are flaviviruses transmitted by mosquitos. There are approximately 390 million DENV infections every year, making dengue virus a major global public health concern. While there is a recently licensed DENV vaccine, it has low efficacy against preventing DENV2 infections. Individuals that are naturally infected with DENV2 generate neutralizing antibodies that can be protective against reinfection with DENV2. By studying three of these neutralizing antibodies, we found that they bind to two different locations on the surface of the virus. Additionally we found that most individuals that were naturally infected with DENV2, have antibodies circulating in their blood that target both of these regions. People who were vaccinated against DENV2 also make antibodies targeting both of these sites, suggesting they might also be protected against DENV2 infection. These studies reveal that human antibodies against DENV2 target the same two regions across multiple individuals. Additionally, for a DENV2 vaccine to be protective, it may be important to elicit antibodies directed to these regions as well.
Collapse
Affiliation(s)
- Emily N. Gallichotte
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Thomas J. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, North Carolina, United States of America
| | - Boyd L. Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, North Carolina, United States of America
| | - Douglas G. Widman
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, North Carolina, United States of America
| | - Anna Durbin
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Steve Whitehead
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- Department of Epidemiology, University of North Carolina at Chapel Hill School of Public Health, Chapel Hill, North Carolina, United States of America
- * E-mail: (RSB); (AMdS)
| | - Aravinda M. de Silva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, United States of America
- * E-mail: (RSB); (AMdS)
| |
Collapse
|
33
|
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: 46] [Impact Index Per Article: 7.7] [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
|
34
|
Rothan HA, Bidokhti MRM, Byrareddy SN. Current concerns and perspectives on Zika virus co-infection with arboviruses and HIV. J Autoimmun 2018; 89:11-20. [PMID: 29352633 DOI: 10.1016/j.jaut.2018.01.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/30/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Abstract
Dissemination of vector-borne viruses, such as Zika virus (ZIKV), in tropical and sub-tropical regions has a complicated impact on the immunopathogenesis of other endemic viruses such as dengue virus (DENV), chikungunya virus (CHIKV) and human immunodeficiency virus (HIV). The consequences of the possible co-infections with these viruses have specifically shown significant impact on the treatment and vaccination strategies. ZIKV is a mosquito-borne flavivirus from African and Asian lineages that causes neurological complications in infected humans. Many of DENV and CHIKV endemic regions have been experiencing outbreaks of ZIKV infection. Intriguingly, the mosquitoes, Aedes Aegypti and Aedes Albopictus, can simultaneously transmit all the combinations of ZIKV, DENV, and CHIKV to the humans. The co-circulation of these viruses leads to a complicated immune response due to the pre-existence or co-existence of ZIKV infection with DENV and CHIKV infections. The non-vector transmission of ZIKV, especially, via sexual intercourse and placenta represents an additional burden that may hander the treatment strategies of other sexually transmitted diseases such as HIV. Collectively, ZIKV co-circulation and co-infection with other viruses have inevitable impact on the host immune response, diagnosis techniques, and vaccine development strategies for the control of these co-infections.
Collapse
Affiliation(s)
- Hussin A Rothan
- Department of Human Biology, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia.
| | - Mehdi R M Bidokhti
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Centre (UNMC), Omaha, NE 68198-5800, USA.
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Centre (UNMC), Omaha, NE 68198-5800, USA.
| |
Collapse
|
35
|
Zika virus structural biology and progress in vaccine development. Biotechnol Adv 2018; 36:47-53. [DOI: 10.1016/j.biotechadv.2017.09.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 01/01/2023]
|