1
|
Porier DL, Adam A, Kang L, Michalak P, Tupik J, Santos MA, Tanelus M, López K, Auguste DI, Lee C, Allen IC, Wang T, Auguste AJ. Humoral and T-cell-mediated responses to an insect-specific flavivirus-based Zika virus vaccine candidate. PLoS Pathog 2024; 20:e1012566. [PMID: 39388457 PMCID: PMC11495591 DOI: 10.1371/journal.ppat.1012566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 10/22/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Flaviviruses represent a significant global health threat and relatively few licensed vaccines exist to protect against them. Insect-specific flaviviruses (ISFVs) are incapable of replication in humans and have emerged as a novel and promising tool for flavivirus vaccine development. ISFV-based flavivirus vaccines have shown exceptional safety, immunogenicity, and efficacy, however, a detailed assessment of the correlates of protection and immune responses induced by these vaccines are still needed for vaccine optimization. Here, we explore the mechanisms of protective immunity induced by a previously created pre-clinical Zika virus (ZIKV) vaccine candidate, called Aripo/Zika (ARPV/ZIKV). In brief, immunocompromised IFN-αβR-/- mice passively immunized with ARPV/ZIKV immune sera experienced protection after lethal ZIKV challenge, although this protection was incomplete. ARPV/ZIKV-vaccinated IFN-αβR-/- mice depleted of CD4+ or CD8+ T-cells at the time of ZIKV challenge showed no morbidity or mortality. However, the adoptive transfer of ARPV/ZIKV-primed T-cells into recipient IFN-αβR-/- mice resulted in a two-day median increase in survival time compared to controls. Altogether, these results suggest that ARPV/ZIKV-induced protection is primarily mediated by neutralizing antibodies at the time of challenge and that T-cells may play a comparatively minor but cumulative role in the protection observed. Lastly, ARPV/ZIKV-vaccinated Tcra KO mice, which are deficient in T-cell responses, experienced significant mortality post-challenge. These results suggest that ARPV/ZIKV-induced cell-mediated responses are critical for development of protective immune responses at vaccination. Despite the strong focus on neutralizing antibody responses to novel flavivirus vaccine candidates, these results suggest that cell-mediated responses induced by ISFV-based vaccines remain important to overall protective responses.
Collapse
Affiliation(s)
- Danielle L. Porier
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lin Kang
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, United States of America
| | - Pawel Michalak
- Department of Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Juselyn Tupik
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
| | - Matthew A. Santos
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Manette Tanelus
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Krisangel López
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Dawn I. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christy Lee
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Irving C. Allen
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Albert J. Auguste
- Department of Entomology, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| |
Collapse
|
2
|
Lingemann M, Amaro-Carambot E, Lamirande EW, Pierson TC, Whitehead SS. Simultaneous quantitation of neutralizing antibodies against all four dengue virus serotypes using optimized reporter virus particles. J Virol 2024; 98:e0068124. [PMID: 38953379 PMCID: PMC11265411 DOI: 10.1128/jvi.00681-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
Serum-neutralizing antibody titers are a critical measure of vaccine immunogenicity and are used to determine flavivirus seroprevalence in study populations. An effective dengue virus (DENV) vaccine must confer simultaneous protection against viruses grouped within four antigenic serotypes. Existing flavivirus neutralization assays, including the commonly used plaque/focus reduction neutralization titer (PRNT/FRNT) assay, require an individual assay for each virus, serotype, and strain and easily become a labor-intensive and time-consuming effort for large epidemiological studies or vaccine trials. Here, we describe a multiplex reporter virus particle neutralization titer (TetraPlex RVPNT) assay for DENV that allows simultaneous quantitative measures of antibody-mediated neutralization of infection against all four DENV serotypes in a single low-volume clinical sample and analyzed by flow cytometry. Comparative studies confirm that the neutralization titers of antibodies measured by the TetraPlex RVPNT assay are similar to FRNT/PRNT assay approaches performed separately for each viral strain. The use of this high-throughput approach enables the careful serological study in DENV endemic populations and vaccine recipients required to support the development of a safe and effective tetravalent DENV vaccine. IMPORTANCE As a mediator of protection against dengue disease and a serological indicator of prior infection, the detection and quantification of neutralizing antibodies against DENV is an important "gold standard" tool. However, execution of traditional neutralizing antibody assays is often cumbersome and requires repeated application for each virus or serotype. The optimized RVPNT assay described here is high-throughput, easily multiplexed across multiple serotypes, and targets reporter viral particles that can be robustly produced for all four DENV serotypes. The use of this transformative RVPNT assay will support the expansion of neutralizing antibody datasets to answer research and public health questions often limited by the more cumbersome neutralizing antibody assays and the need for greater quantities of test serum.
Collapse
Affiliation(s)
- Matthias Lingemann
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Translational Immunobiology Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Emérito Amaro-Carambot
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine W. Lamirande
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Theodore C. Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Arbovirus Immunology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen S. Whitehead
- Arbovirus Vaccine Research Section, Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Ackermann-Gäumann R, Lang P, Zens KD. Defining the "Correlate(s) of Protection" to tick-borne encephalitis vaccination and infection - key points and outstanding questions. Front Immunol 2024; 15:1352720. [PMID: 38318179 PMCID: PMC10840404 DOI: 10.3389/fimmu.2024.1352720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/05/2024] [Indexed: 02/07/2024] Open
Abstract
Tick-borne Encephalitis (TBE) is a severe disease of the Central Nervous System (CNS) caused by the tick-borne encephalitis virus (TBEV). The generation of protective immunity after TBEV infection or TBE vaccination relies on the integrated responses of many distinct cell types at distinct physical locations. While long-lasting memory immune responses, in particular, form the basis for the correlates of protection against many diseases, these correlates of protection have not yet been clearly defined for TBE. This review addresses the immune control of TBEV infection and responses to TBE vaccination. Potential correlates of protection and the durability of protection against disease are discussed, along with outstanding questions in the field and possible areas for future research.
Collapse
Affiliation(s)
- Rahel Ackermann-Gäumann
- Microbiologie, ADMED Analyses et Diagnostics Médicaux, La Chaux-de-Fonds, Switzerland
- Swiss National Reference Center for Tick-transmitted Diseases, La Chaux-de-Fonds, Switzerland
| | - Phung Lang
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
| | - Kyra D. Zens
- Epidemiology, Biostatistics and Prevention Institute, University of Zurich, Zurich, Switzerland
- Institute for Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Bos S, Graber AL, Cardona-Ospina JA, Duarte EM, Zambrana JV, Ruíz Salinas JA, Mercado-Hernandez R, Singh T, Katzelnick LC, de Silva A, Kuan G, Balmaseda A, Harris E. Protection against symptomatic dengue infection by neutralizing antibodies varies by infection history and infecting serotype. Nat Commun 2024; 15:382. [PMID: 38195666 PMCID: PMC10776616 DOI: 10.1038/s41467-023-44330-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/08/2023] [Indexed: 01/11/2024] Open
Abstract
Dengue viruses (DENV1-4) are the most prevalent arboviruses in humans and a major public health concern. Understanding immune mechanisms that modulate DENV infection outcome is critical for vaccine development. Neutralizing antibodies (nAbs) are an essential component of the protective immune response, yet their measurement often relies on a single cellular substrate and partially mature virions, which does not capture the full breadth of neutralizing activity and may lead to biased estimations of nAb potency. Here, we analyze 125 samples collected after one or more DENV infections but prior to subsequent symptomatic or inapparent DENV1, DENV2, or DENV3 infections from a long-standing pediatric cohort study in Nicaragua. By assessing nAb responses using Vero cells with or without DC-SIGN and with mature or partially mature virions, we find that nAb potency and the protective NT50 cutoff are greatly influenced by cell substrate and virion maturation state. Additionally, the correlation between nAb titer and protection from disease depends on prior infection history and infecting serotype. Finally, we uncover variations in nAb composition that contribute to protection from symptomatic infection differently after primary and secondary prior infection. These findings have important implications for identifying antibody correlates of protection for vaccines and natural infections.
Collapse
Affiliation(s)
- Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| | - Aaron L Graber
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Jaime A Cardona-Ospina
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Grupo de Investigación Biomedicina, Facultad de Medicina, Institución Universitaria Visión de las Américas, Pereira, Colombia
| | - Elias M Duarte
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Jose Victor Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | - Reinaldo Mercado-Hernandez
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Tulika Singh
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Leah C Katzelnick
- Viral Epidemiology and Immunity Unit, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Aravinda de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Guillermina Kuan
- Sustainable Sciences Institute, Managua, Nicaragua
- Centro de Salud Sócrates Flores Vivas, Ministerio de Salud, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnóstico y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
5
|
Raynes JM, Young PG, Lorenz N, Loh JM, McGregor R, Baker EN, Proft T, Moreland NJ. Identification of an immunodominant region on a group A Streptococcus T-antigen reveals temperature-dependent motion in pili. Virulence 2023; 14:2180228. [PMID: 36809931 PMCID: PMC9980535 DOI: 10.1080/21505594.2023.2180228] [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] [Indexed: 02/24/2023] Open
Abstract
Group A Streptococcus (GAS) is a globally important pathogen causing a broad range of human diseases. GAS pili are elongated proteins with a backbone comprised repeating T-antigen subunits, which extend from the cell surface and have important roles in adhesion and establishing infection. No GAS vaccines are currently available, but T-antigen-based candidates are in pre-clinical development. This study investigated antibody-T-antigen interactions to gain molecular insight into functional antibody responses to GAS pili. Large, chimeric mouse/human Fab-phage libraries generated from mice vaccinated with the complete T18.1 pilus were screened against recombinant T18.1, a representative two-domain T-antigen. Of the two Fab identified for further characterization, one (designated E3) was cross-reactive and also recognized T3.2 and T13, while the other (H3) was type-specific reacting with only T18.1/T18.2 within a T-antigen panel representative of the major GAS T-types. The epitopes for the two Fab, determined by x-ray crystallography and peptide tiling, overlapped and mapped to the N-terminal region of the T18.1 N-domain. This region is predicted to be buried in the polymerized pilus by the C-domain of the next T-antigen subunit. However, flow cytometry and opsonophagocytic assays showed that these epitopes were accessible in the polymerized pilus at 37°C, though not at lower temperature. This suggests that there is motion within the pilus at physiological temperature, with structural analysis of a covalently linked T18.1 dimer indicating "knee-joint" like bending occurs between T-antigen subunits to expose this immunodominant region. This temperature dependent, mechanistic flexing provides new insight into how antibodies interact with T-antigens during infection.
Collapse
Affiliation(s)
- Jeremy M. Raynes
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Paul G. Young
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand,School of Biological Sciences, The University of Auckland, Auckland, New Zealand,CONTACT Paul G. Young
| | - Natalie Lorenz
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Jacelyn M.S. Loh
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Reuben McGregor
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Edward N. Baker
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand,School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| | - Thomas Proft
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Nicole J. Moreland
- School of Medical Sciences, The University of Auckland, Auckland, New Zealand,Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand,Nicole J. Moreland
| |
Collapse
|
6
|
Munt JE, Henein S, Adams C, Young E, Hou YJ, Conrad H, Zhu D, Dong S, Kose N, Yount B, Meganck RM, Tse LPV, Kuan G, Balmaseda A, Ricciardi MJ, Watkins DI, Crowe JE, Harris E, DeSilva AM, Baric RS. Homotypic antibodies target novel E glycoprotein domains after natural DENV 3 infection/vaccination. Cell Host Microbe 2023; 31:1850-1865.e5. [PMID: 37909048 PMCID: PMC11221912 DOI: 10.1016/j.chom.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 07/31/2023] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
The envelope (E) glycoprotein is the primary target of type-specific (TS) neutralizing antibodies (nAbs) after infection with any of the four distinct dengue virus serotypes (DENV1-4). nAbs can be elicited to distinct structural E domains (EDs) I, II, or III. However, the relative contribution of these domain-specific antibodies is unclear. To identify the primary DENV3 nAb targets in sera after natural infection or vaccination, chimeric DENV1 recombinant encoding DENV3 EDI, EDII, or EDIII were generated. DENV3 EDII is the principal target of TS polyclonal nAb responses and encodes two or more neutralizing epitopes. In contrast, some were individuals vaccinated with a DENV3 monovalent vaccine-elicited serum TS nAbs targeting each ED in a subject-dependent fashion, with an emphasis on EDI and EDIII. Vaccine responses were also sensitive to DENV3 genotypic variation. This DENV1/3 panel allows the measurement of serum ED TS nAbs, revealing differences in TS nAb immunity after natural infection or vaccination.
Collapse
Affiliation(s)
- Jennifer E Munt
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Sandra Henein
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Cameron Adams
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Ellen Young
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Yixuan J Hou
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Helen Conrad
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Deanna Zhu
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Stephanie Dong
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Nashville, TN, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Rita M Meganck
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Long Ping V Tse
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Guillermina Kuan
- Health Center Socrates Flores Vivas, Ministry of Health, Managua, Nicaragua; Sustainable Sciences Institute, Managua, Nicaragua
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua; National Virology Laboratory, National Center for Diagnosis and Reference, Ministry of Health, Managua, Nicaragua
| | | | - David I Watkins
- University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, CA, USA
| | - Aravinda M DeSilva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Bos S, Graber AL, Cardona-Ospina JA, Duarte EM, Zambrana JV, Ruíz Salinas JA, Mercado-Hernandez R, Singh T, Katzelnick LC, de Silva A, Kuan G, Balmaseda A, Harris E. The association of neutralizing antibodies with protection against symptomatic dengue virus infection varies by serotype, prior infection history, and assay condition. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.20.23291522. [PMID: 37502957 PMCID: PMC10371115 DOI: 10.1101/2023.06.20.23291522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The four dengue virus serotypes (DENV1-4) are the most prevalent arboviruses in humans and a major public health concern worldwide. Understanding immune mechanisms that modulate DENV infection outcome is critical for epidemic preparedness and development of a safe and effective vaccine. Neutralizing antibodies (nAbs) are an essential component of the protective response, yet their measurement often relies on a single cellular substrate and partially mature virions, which do not capture the full breadth of neutralizing activity and may lead to biased estimations of nAb potency. Here, we investigated the characteristics of nAbs associated with protection against dengue cases using samples collected after one or more DENV infections but prior to subsequent symptomatic or inapparent DENV1, DENV2, or DENV3 infections from a long- standing pediatric cohort study in Nicaragua. By assessing nAb responses using Vero cells with or without the attachment factor DC-SIGN and with mature or partially mature virions, we found that nAb potency and the protective NT 50 cutoff were greatly influenced by cell substrate and virion maturation state. Additionally, the correlation between nAb titer and protection from disease depended on an individual's prior infection history and the subsequent infecting DENV serotype. Finally, we uncovered variations in nAbs composition that contributed to protection from symptomatic DENV infection differently after primary and secondary prior infection. These findings have important implications for identifying antibody correlates of protection in the context of vaccines and natural infections.
Collapse
|
8
|
LeFevre I, Bravo L, Folschweiller N, Medina EL, Moreira ED, Nordio F, Sharma M, Tharenos LM, Tricou V, Watanaveeradej V, Winkle PJ, Biswal S. Bridging the immunogenicity of a tetravalent dengue vaccine (TAK-003) from children and adolescents to adults. NPJ Vaccines 2023; 8:75. [PMID: 37230978 DOI: 10.1038/s41541-023-00670-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 05/10/2023] [Indexed: 05/27/2023] Open
Abstract
Immunobridging is an important methodology that can be used to extrapolate vaccine efficacy estimates to populations not evaluated in clinical studies, and that has been successfully used in developing many vaccines. Dengue, caused by a mosquito-transmitted flavivirus endemic to many tropical and subtropical regions, is traditionally thought of as a pediatric disease but is now a global threat to both children and adults. We bridged immunogenicity data from a phase 3 efficacy study of a tetravalent dengue vaccine (TAK-003), performed in children and adolescents living in endemic areas, with an immunogenicity study in adults in non-endemic areas. Neutralizing antibody responses were comparable in both studies following receipt of a two-dose TAK-003 schedule (months 0 and 3). Similar immune responses were observed across exploratory assessments of additional humoral responses. These data support the potential for clinical efficacy of TAK-003 in adults.
Collapse
Affiliation(s)
- Inge LeFevre
- Vaccines Business Unit, Takeda Pharmaceuticals International AG, Zürich, Switzerland
| | - Lulu Bravo
- College of Medicine, University of the Philippines, Manila, Philippines
| | - Nicolas Folschweiller
- Vaccines Business Unit, Takeda Pharmaceuticals International AG, Zürich, Switzerland
| | - Eduardo Lopez Medina
- Centro de Estudios en Infectología Pediatrica CEIP; Department of Pediatrics, Universidad Del Valle; Clínica Imbanaco, Grupo Quironsalud, Cali, Colombia
| | - Edson Duarte Moreira
- Associação Obras Sociais Irmã Dulce Hospital Santo Antônio and Oswaldo Cruz Foundation, Bahia, Brazil
| | | | | | - Leslie M Tharenos
- The Division of Environmental and Occupational Health Sciences, University of Illinois at Chicago School of Public Health, Chicago, IL, USA
| | - Vianney Tricou
- Vaccines Business Unit, Takeda Pharmaceuticals International AG, Zürich, Switzerland
| | - Veerachai Watanaveeradej
- Department of Pediatrics, Phramongkutklao Hospital and Faculty of Medicine, Kasetsart University, Bangkok, Thailand
| | | | | |
Collapse
|
9
|
Kubinski M, Beicht J, Zdora I, Biermann J, Puff C, Gerlach T, Tscherne A, Baumgärtner W, Osterhaus ADME, Sutter G, Prajeeth CK, Rimmelzwaan GF. A recombinant Modified Vaccinia virus Ankara expressing prME of tick-borne encephalitis virus affords mice full protection against TBEV infection. Front Immunol 2023; 14:1182963. [PMID: 37153588 PMCID: PMC10160477 DOI: 10.3389/fimmu.2023.1182963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Tick-borne encephalitis virus (TBEV) is an important human pathogen that can cause a serious disease involving the central nervous system (tick-borne encephalitis, TBE). Although approved inactivated vaccines are available, the number of TBE cases is rising, and breakthrough infections in fully vaccinated subjects have been reported in recent years. Methods In the present study, we generated and characterized a recombinant Modified Vaccinia virus Ankara (MVA) for the delivery of the pre-membrane (prM) and envelope (E) proteins of TBEV (MVA-prME). Results MVA-prME was tested in mice in comparison with a licensed vaccine FSME-IMMUN® and proved to be highly immunogenic and afforded full protection against challenge infection with TBEV. Discussion Our data indicate that MVA-prME holds promise as an improved next-generation vaccine for the prevention of TBE.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Isabel Zdora
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, Hannover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hannover, Germany
| | - Jeannine Biermann
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Christina Puff
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Alina Tscherne
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- Center for Systems Neuroscience, Hannover Graduate School for Neurosciences, Infection Medicine, and Veterinary Sciences (HGNI), Hannover, Germany
| | - Albert D. M. E. Osterhaus
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Gerd Sutter
- Division of Virology, Institute for Infectious Diseases and Zoonoses, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
- *Correspondence: Guus F. Rimmelzwaan,
| |
Collapse
|
10
|
Nygren TM, Pilic A, Böhmer MM, Wagner-Wiening C, Wichmann O, Harder T, Hellenbrand W. Tick-borne encephalitis vaccine effectiveness and barriers to vaccination in Germany. Sci Rep 2022; 12:11706. [PMID: 35810184 PMCID: PMC9271034 DOI: 10.1038/s41598-022-15447-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Tick-borne encephalitis (TBE) vaccination coverage remains low in Germany. Our case-control study (2018-2020) aimed to examine reasons for low vaccine uptake, vaccine effectiveness (VE), and vaccine breakthrough infections (VBIs). Telephone interviews (581 cases, 975 matched controls) covered vaccinations, vaccination barriers, and confounders identified with directed acyclic graphs. Multivariable logistic regression determined VE as 1-odds ratio with 95% confidence intervals (CI). We additionally calculated VE with the Screening method using routine surveillance and vaccination coverage data. Main vaccination barriers were poor risk perception and fear of adverse events. VE was 96.6% (95% CI 93.7-98.2) for ≥ 3 doses and manufacturer-recommended dosing intervals. Without boosters, VE after ≥ 3 doses at ≤ 10 years was 91.2% (95% CI 82.7-95.6). VE was similar for homologous/heterologous vaccination. Utilising routine surveillance data, VE was comparable (≥ 3 doses: 92.8%). VBIs (n = 17, 2.9% of cases) were older, had more comorbidities and higher severity than unvaccinated cases. However, only few VBIs were diagnostically confirmed; 57% of re-tested vaccinated cases (≥ 1 dose, n = 54) proved false positive. To increase TBE vaccine uptake, communication efforts should address complacency and increase confidence in the vaccines' safety. The observed duration of high VE may inform decision-makers to consider extending booster intervals to 10 years.
Collapse
Affiliation(s)
- Teresa M Nygren
- Immunisation Unit, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany.
- Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Antonia Pilic
- Immunisation Unit, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| | - Merle M Böhmer
- Bavarian Health and Food Safety Authority (LGL), Oberschleißheim, Germany
- Institute of Social Medicine and Health Systems Research, Otto-Von-Guericke-University Magdeburg, Magdeburg, Germany
| | | | - Ole Wichmann
- Immunisation Unit, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| | - Thomas Harder
- Immunisation Unit, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| | - Wiebke Hellenbrand
- Immunisation Unit, Robert Koch Institute, Seestraße 10, 13353, Berlin, Germany
| |
Collapse
|
11
|
Panchal R, Bapat S, Mukherjee S, Chowdhary A. In silico binding analysis of lutein and rosmarinic acid against envelope domain III protein of dengue virus. Indian J Pharmacol 2021; 53:471-479. [PMID: 34975135 PMCID: PMC8764985 DOI: 10.4103/ijp.ijp_576_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 08/29/2020] [Accepted: 11/11/2021] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVE The study was performed to evaluate in silico binding ability of lutein and rosmarinic acid (RA) with the envelope domain III (EDIII) proteins of the four serotypes of dengue virus (DENV), enlightening potential antiviral activity of the two compounds. MATERIALS AND METHODS EDIII protein structures for the four DENV serotypes were retrieved from RCSB Protein data bank (PDB) and used as receptors. Four ligands of lutein and four of RA were selected from the ZINC database and used for computational molecular docking and ligand interaction analysis with the four receptors using bioinformatics tools like AutoDock Vina and Molecular Operating Environment (MOE) software. RESULTS The EDIII of the four serotypes demonstrated significant interaction with ligands of lutein and RA. RA ligand ZINC899870, particularly presented best-binding energy values of 6.4, -7.0, and 6.9 kcal/mol with EDIII of serotype DENV-1, DENV-2, and DENV-4 respectively. Whereas, lutein ligand, ZINC14879959 presented best-binding energy value of 7.9 kcal/mol for EDIII of serotype DENV-3. From the results predicted by MOE, the hydroxyl (OH) of 3, 4-dihydroxyphenyl group of RA ligand ZINC899870 is actively involved in interaction with all four serotypes. CONCLUSION RA is a competent candidate for further evaluation of potential in vitro antiviral activity that can be effective in conferring protection against the four serotypes of DENV.
Collapse
Affiliation(s)
- Ritesh Panchal
- School of Science, Narsee Monjee Institute of Management Studies, Mumbai, India
| | - Sanket Bapat
- MIT School of Bioengineering Science and Research, ADT University, Pune, India
| | - Sandeepan Mukherjee
- Department of Virology, Haffkin Institute for Training, Research and Testing, Mumbai, India
| | - Abhay Chowdhary
- Department of Microbiology, D. Y. Patil School of Medicine, Navi Mumbai, India
| |
Collapse
|
12
|
Qu S, Wang X, Yang L, Meng R, Feng C, Yang B, Huang J, Li Q, Wang J, Zhang D. Mapping of a unique epitope on domain III of the envelope protein of Tembusu virus. Virus Res 2021; 306:198582. [PMID: 34599934 DOI: 10.1016/j.virusres.2021.198582] [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: 06/26/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
We recently developed a Tembusu virus (TMUV)-specific monoclonal antibody (MAb) 12F11, which was found to recognize a long amino acid sequence between residues 8 and 77 of domain III of the envelope protein (EDIII). Here, the epitope recognized by MAb 12F11 was mapped using alanine substitutions combined with dissociation constant analysis. The findings, and prediction of tertiary structure of TMUV EDIII, showed that the MAb 12F11 epitope contained one critical residue and 13 peripheral residues. Moreover, the antigenic site was shown to span four loops (N-terminal region, AB, BC, and CD) and three β-strands (A, B, and D). The present work contributes to the understanding of antigenic structure of TMUV envelope protein.
Collapse
Affiliation(s)
- Shenghua Qu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Chonglun Feng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Baolin Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Jingjing Huang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Qiong Li
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Jiaying Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China.
| |
Collapse
|
13
|
Are the Organoid Models an Invaluable Contribution to ZIKA Virus Research? Pathogens 2021; 10:pathogens10101233. [PMID: 34684182 PMCID: PMC8537471 DOI: 10.3390/pathogens10101233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/15/2021] [Accepted: 09/20/2021] [Indexed: 12/16/2022] Open
Abstract
In order to prevent new pathogen outbreaks and avoid possible new global health threats, it is important to study the mechanisms of microbial pathogenesis, screen new antiviral agents and test new vaccines using the best methods. In the last decade, organoids have provided a groundbreaking opportunity for modeling pathogen infections in human brains, including Zika virus (ZIKV) infection. ZIKV is a member of the Flavivirus genus, and it is recognized as an emerging infectious agent and a serious threat to global health. Organoids are 3D complex cellular models that offer an in-scale organ that is physiologically alike to the original one, useful for exploring the mechanisms behind pathogens infection; additionally, organoids integrate data generated in vitro with traditional tools and often support those obtained in vivo with animal model. In this mini-review the value of organoids for ZIKV research is examined and sustained by the most recent literature. Within a 3D viewpoint, tissue engineered models are proposed as future biological systems to help in deciphering pathogenic processes and evaluate preventive and therapeutic strategies against ZIKV. The next steps in this field constitute a challenge that may protect people and future generations from severe brain defects.
Collapse
|
14
|
Fumagalli MJ, Figueiredo LTM, Aquino VH. Linear and Continuous Flavivirus Epitopes From Naturally Infected Humans. Front Cell Infect Microbiol 2021; 11:710551. [PMID: 34458161 PMCID: PMC8387565 DOI: 10.3389/fcimb.2021.710551] [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: 05/16/2021] [Accepted: 07/15/2021] [Indexed: 12/27/2022] Open
Abstract
This manuscript is an up-to-date review of experimentally validated linear and continuous epitopes identified from arbovirus members of the Flavivirus genus. We summarized 153 immunoreactive peptides from the Dengue virus, Zika virus, Japanese encephalitis virus, West Nile virus, and tick-borne encephalitis virus described in studies published from 1989 to 2020. We included peptides from structural (envelope, capsid, and pre-membrane) and nonstructural (Ns1–5) viral proteins that demonstrated relevant immunoreactivity with antibodies from naturally infected or vaccinated humans. We included peptides that demonstrated relevant reactivity features, such as indicators of disease severity related to immunological or immunopathological outcomes, differential or group diagnostic markers, immunotherapy candidates, and potential for vaccine formulation. The majority of immunoreactive peptides were described for DENV probably due to its long-lasting impact on human health and the lack of efficient vaccines and therapeutic methods. Immune landscape data regarding linear immunoreactive and continuous flavivirus peptides are still scarce, and a complete and more detailed map remains to be elucidated. Therefore, this review provides valuable data for those investigating the antibody response against flavivirus infection.
Collapse
Affiliation(s)
- Marcilio Jorge Fumagalli
- Virology Research Center, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Victor Hugo Aquino
- Laboratory of Virology, Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Substantial Attenuation of Virulence of Tembusu Virus Strain PS Is Determined by an Arginine at Residue 304 of the Envelope Protein. J Virol 2021; 95:JVI.02331-20. [PMID: 33328312 DOI: 10.1128/jvi.02331-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
The Tembusu virus (TMUV) PS strain, derived by several passages and plaque purifications in BHK-21 cells, displays markedly lower virulence in Pekin ducklings relative to a natural isolate of TMUV, but the potential virulence determinants and the in vivo mechanisms for substantial virulence attenuation of the passage variant remain unknown. Here, we constructed a series of chimeric and mutant viruses and assessed their virulence using a 2-day-old Pekin duckling model. We showed that residue 304 in the envelope (E) protein is the molecular determinant of TMUV virulence. Further investigations with mutant and parental viruses demonstrated that acquisition of positive charges at E protein residue 304 plays a critical role in substantial attenuation of neurovirulence and neuroinvasiveness, which is linked to enhanced binding affinity for glycosaminoglycans (GAGs). In Pekin ducklings infected by subcutaneous inoculation, an Arg at residue 304 in the E protein was shown to contribute to more rapid virus clearance from the circulation, markedly reduced viremia, and significantly decreased viral growth in the extraneural tissues and the central nervous system, relative to a Met at the corresponding residue. These findings suggest that the in vivo mechanism of virulence attenuation of the TMUV passage variant closely resembles that proposed previously for GAG-binding variants of other flaviviruses. Overall, our study provides insight into the molecular basis of TMUV virulence and the in vivo consequences of acquisition of a GAG-binding determinant at residue 304 in the E protein of TMUV.IMPORTANCE TMUV-related disease emerged in 2010 and has a significant economic impact on the duck industry. Although the disease was originally recognized to affect adult ducks, increasing evidence has shown that TMUV also causes severe disease of young ducklings. It is, therefore, essential to investigate the pathogenesis of TMUV infection in a young duckling model. The significance of our studies is in identifying E protein residue Arg304 as the molecular determinant for TMUV virulence and in clarifying the crucial role of positive charges at E protein residue 304 in virulence attenuation of a TMUV passage variant. These data will greatly enhance our understanding of the pathogenesis of TMUV infection in ducklings and have implications for development of a safe and efficient vaccine.
Collapse
|
16
|
Zhang Y, Wang S, Wu Y, Hou W, Yuan L, Shen C, Wang J, Ye J, Zheng Q, Ma J, Xu J, Wei M, Li Z, Nian S, Xiong H, Zhang L, Shi Y, Fu B, Cao J, Yang C, Li Z, Yang T, Liu L, Yu H, Hu J, Ge S, Chen Y, Zhang T, Zhang J, Cheng T, Yuan Q, Xia N. Virus-Free and Live-Cell Visualizing SARS-CoV-2 Cell Entry for Studies of Neutralizing Antibodies and Compound Inhibitors. SMALL METHODS 2021; 5:2001031. [PMID: 33614907 PMCID: PMC7883248 DOI: 10.1002/smtd.202001031] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/25/2020] [Indexed: 05/07/2023]
Abstract
The ongoing corona virus disease 2019 (COVID-19) pandemic, caused by SARS-CoV-2 infection, has resulted in hundreds of thousands of deaths. Cellular entry of SARS-CoV-2, which is mediated by the viral spike protein and ACE2 receptor, is an essential target for the development of vaccines, therapeutic antibodies, and drugs. Using a mammalian cell expression system, a genetically engineered sensor of fluorescent protein (Gamillus)-fused SARS-CoV-2 spike trimer (STG) to probe the viral entry process is developed. In ACE2-expressing cells, it is found that the STG probe has excellent performance in the live-cell visualization of receptor binding, cellular uptake, and intracellular trafficking of SARS-CoV-2 under virus-free conditions. The new system allows quantitative analyses of the inhibition potentials and detailed influence of COVID-19-convalescent human plasmas, neutralizing antibodies and compounds, providing a versatile tool for high-throughput screening and phenotypic characterization of SARS-CoV-2 entry inhibitors. This approach may also be adapted to develop a viral entry visualization system for other viruses.
Collapse
Affiliation(s)
- Yali Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Shaojuan Wang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Yangtao Wu
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Wangheng Hou
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Lunzhi Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chenguang Shen
- Shenzhen Key Laboratory of Pathogen and ImmunityNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalSecond Hospital Affiliated to Southern University of Science and TechnologyShenzhenGuangdong518112China
| | - Juan Wang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jianghui Ye
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Qingbing Zheng
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jian Ma
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jingjing Xu
- Department of HematologyFujian Medical University Union HospitalFujian Provincial Key Laboratory on HematologyFujian Institute of HematologyFuzhouFujian350001China
| | - Min Wei
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zonglin Li
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Sheng Nian
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Hualong Xiong
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Liang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Yang Shi
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Baorong Fu
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jiali Cao
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Chuanlai Yang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Zhiyong Li
- The First Hospital of Xiamen UniversityXiamen361003China
| | - Ting Yang
- Department of HematologyFujian Medical University Union HospitalFujian Provincial Key Laboratory on HematologyFujian Institute of HematologyFuzhouFujian350001China
| | - Lei Liu
- Shenzhen Key Laboratory of Pathogen and ImmunityNational Clinical Research Center for Infectious DiseaseShenzhen Third People's HospitalSecond Hospital Affiliated to Southern University of Science and TechnologyShenzhenGuangdong518112China
| | - Hai Yu
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jianda Hu
- Department of HematologyFujian Medical University Union HospitalFujian Provincial Key Laboratory on HematologyFujian Institute of HematologyFuzhouFujian350001China
| | - Shengxiang Ge
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Yixin Chen
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesSchool of Public Health & School of Life SciencesXiamen UniversityXiamenFujian361102China
| |
Collapse
|
17
|
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: 3.5] [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
|
18
|
Post-Vaccination Yellow Fever Antiserum Reduces Zika Virus in Embryoid Bodies When Placental Cells are Present. Vaccines (Basel) 2020; 8:vaccines8040752. [PMID: 33322247 PMCID: PMC7768546 DOI: 10.3390/vaccines8040752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/09/2020] [Accepted: 11/29/2020] [Indexed: 11/18/2022] Open
Abstract
Zika virus (ZIKV) is a flavivirus that originated in Africa but emerged in Latin America in 2015. In this region, other flaviviruses such as Dengue (DENV), West Nile, and Yellow Fever virus (YFV) also circulate, allowing for possible antigenic cross-reactivity to impact viral infections and immune responses. Studies have found antibody-mediated enhancement between DENV and ZIKV, but the impact of YFV antibodies on ZIKV infection has not been fully explored. ZIKV infections cause congenital syndromes, such as microcephaly, necessitating further research into ZIKV vertical transmission through the placental barrier. Recent advancements in biomedical engineering have generated co-culture methods that allow for the in vitro recapitulation of the maternal–fetal interface. This study utilized a transwell assay, which was a co-culture model utilizing human placental syncytiotrophoblasts, fetal umbilical cells, and a differentiating embryoid body, to replicate the maternal–fetal axis. To determine if cross-reactive YFV vaccine antibodies impacted the pathogenesis of ZIKV across the maternal–fetal axis, syncytiotrophoblasts were inoculated with ZIKV or ZIKV incubated with YFV vaccine antisera, and the viral load was measured 72 h post-inoculation. Here, we report that BeWo and HUVEC cells were permissive to ZIKV and that the impact of YFV post-vaccination antibodies on ZIKV replication was cell line-dependent. Embryoid bodies were also permissive to ZIKV, and the presence of YFV antibodies collected 4–14 months post-vaccination reduced ZIKV infection when placental cells were present. However, when directly infected with ZIKV, the embryoid bodies displayed significantly increased viral loads in the presence of YFV antiserum taken 30 days post-vaccination. The data show that each of the cell lines and EBs have a unique response to ZIKV complexed with post-vaccination serum, suggesting there may be cell-specific mechanisms that impact congenital ZIKV infections. Since ZIKV infections can cause severe congenital syndromes, it is crucial to understand any potential enhancement or protection offered from cross-reactive, post-vaccination antibodies.
Collapse
|
19
|
Merbah M, Wollen-Roberts S, Shubin Z, Li Y, Bai H, Dussupt V, Mendez-Rivera L, Slike B, Krebs SJ, Modjarrad K, Michael NL, Rolland M. A high-throughput multiplex assay to characterize flavivirus-specific immunoglobulins. J Immunol Methods 2020; 487:112874. [PMID: 33022219 DOI: 10.1016/j.jim.2020.112874] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 12/30/2022]
Abstract
Genus Flavivirus, which includes 53 virus species, is the leading cause of arthropod-borne diseases in humans. Diagnosis of these viral diseases is complicated by their overlapping epidemiology and clinical manifestations, and the fact that cross-reactive antibody responses are frequently elicited by individuals in response to infection. We developed a bead-based immunoassay to concomitantly profile the isotype and subclass of antibody responses (five isotypes and four subclasses) in parallel with specificity against multiple antigens. Our panel included 22 envelope (E) and non-structural 1 (NS1) proteins of different flaviviruses (Zika (ZIKV), Dengue (DENV), Yellow Fever (YFV), West Nile (WNV), Japanese Encephalitis (JEV) and Tick-Borne Encephalitis (TBEV)) and the envelope protein of Chikungunya virus (CHIKV). Using 54 samples from 40 individuals with ZIKV infection that had been pre-characterized, we identified 1) stronger ZIKV responses in individuals previously exposed to flavivirus compared to flavivirus-naïve individuals; 2) different antibody isotypes depending on the stage of infection: acute, convalescent and late convalescent; 3) cross-reactive responses; and 4) a potential CHIKV infection. The assay had a broad dynamic range (>5 logs) and has the potential to distinguish antigen-specific responses induced by ZIKV infection from cross-reactive responses. The multidimensional data provided by this high-throughput antibody-profiling platform can advance our understanding of the human immune response to flaviviruses as they expand their global reach.
Collapse
Affiliation(s)
- Mélanie Merbah
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA.
| | - Suzanne Wollen-Roberts
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Zhanna Shubin
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Yifan Li
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Hongjun Bai
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Vincent Dussupt
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Letzibeth Mendez-Rivera
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Bonnie Slike
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Shelly J Krebs
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L Michael
- Center for Infectious Diseases Research, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Morgane Rolland
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| |
Collapse
|
20
|
Kubinski M, Beicht J, Gerlach T, Volz A, Sutter G, Rimmelzwaan GF. Tick-Borne Encephalitis Virus: A Quest for Better Vaccines against a Virus on the Rise. Vaccines (Basel) 2020; 8:E451. [PMID: 32806696 PMCID: PMC7564546 DOI: 10.3390/vaccines8030451] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV), a member of the family Flaviviridae, is one of the most important tick-transmitted viruses in Europe and Asia. Being a neurotropic virus, TBEV causes infection of the central nervous system, leading to various (permanent) neurological disorders summarized as tick-borne encephalitis (TBE). The incidence of TBE cases has increased due to the expansion of TBEV and its vectors. Since antiviral treatment is lacking, vaccination against TBEV is the most important protective measure. However, vaccination coverage is relatively low and immunogenicity of the currently available vaccines is limited, which may account for the vaccine failures that are observed. Understanding the TBEV-specific correlates of protection is of pivotal importance for developing novel and improved TBEV vaccines. For affording robust protection against infection and development of TBE, vaccines should induce both humoral and cellular immunity. In this review, the adaptive immunity induced upon TBEV infection and vaccination as well as novel approaches to produce improved TBEV vaccines are discussed.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany;
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University (LMU) Munich, Veterinaerstr. 13, 80539 Munich, Germany;
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| |
Collapse
|
21
|
The Effects of Pre-Existing Antibodies on Live-Attenuated Viral Vaccines. Viruses 2020; 12:v12050520. [PMID: 32397218 PMCID: PMC7290594 DOI: 10.3390/v12050520] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/11/2022] Open
Abstract
Live-attenuated vaccines (LAVs) have achieved remarkable successes in controlling virus spread, as well as for other applications such as cancer immunotherapy. However, with rapid increases in international travel, globalization, geographic spread of viral vectors, and widespread use of vaccines, there is an increasing need to consider how pre-exposure to viruses which share similar antigenic regions can impact vaccine efficacy. Pre-existing antibodies, derived from either from maternal–fetal transmission, or by previous infection or vaccination, have been demonstrated to interfere with vaccine immunogenicity of measles, adenovirus, and influenza LAVs. Immune interference of LAVs can be caused by the formation of virus–antibody complexes that neutralize virus infection in antigen-presenting cells, or by the cross-linking of the B-cell receptor with the inhibitory receptor, FcγRIIB. On the other hand, pre-existing antibodies can augment flaviviral LAV efficacy such as that of dengue and yellow fever virus, especially when pre-existing antibodies are present at sub-neutralizing levels. The increased vaccine immunogenicity can be facilitated by antibody-dependent enhancement of virus infection, enhancing virus uptake in antigen-presenting cells, and robust induction of innate immune responses that promote vaccine immunogenicity. This review examines the literature on this topic and examines the circumstances where pre-existing antibodies can inhibit or enhance LAV efficacy. A better knowledge of the underlying mechanisms involved could allow us to better manage immunization in seropositive individuals and even identify possibilities that could allow us to exploit pre-existing antibodies to boost vaccine-induced responses for improved vaccine efficacy.
Collapse
|
22
|
Wang L, Wang R, Wang L, Ben H, Yu L, Gao F, Shi X, Yin C, Zhang F, Xiang Y, Zhang L. Structural Basis for Neutralization and Protection by a Zika Virus-Specific Human Antibody. Cell Rep 2020; 26:3360-3368.e5. [PMID: 30893607 DOI: 10.1016/j.celrep.2019.02.062] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 11/22/2018] [Accepted: 02/13/2019] [Indexed: 12/26/2022] Open
Abstract
We previously reported a human monoclonal antibody, ZK2B10, capable of protection against Zika virus (ZIKV) infection and microcephaly in developing mouse embryos. Here, we report the structural features and mechanism of action of ZK2B10. The crystal structure at a resolution of 2.32 Å revealed that the epitope is located on the lateral ridge of DIII of the envelope glycoprotein. Cryo-EM structure with mature ZIKV showed that the antibody binds to DIIIs around the icosahedral 2-fold, 3-fold, and 5-fold axes, a distinct feature compared to those reported for DIII-specific antibodies. The binding of ZK2B10 to ZIKV has no detectable effect on viral attachment to target cells or on conformational changes of the E glycoprotein in the acidic environment, suggesting that ZK2B10 functions at steps between the formation of the fusion intermediate and membrane fusion. These results provide structural and mechanistic insights into how ZK2B10 mediates protection against ZIKV infection.
Collapse
Affiliation(s)
- Lin Wang
- Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Ruoke Wang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lei Wang
- Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haijing Ben
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Fei Gao
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xuanling Shi
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chibiao Yin
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Ye Xiang
- Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Linqi Zhang
- Comprehensive AIDS Research Center, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Advanced Innovation Center for Structural Biology, Center for Global Health and Infectious Diseases, Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
23
|
Lee CYP, Carissimo G, Chen Z, Lum FM, Abu Bakar F, Rajarethinam R, Teo TH, Torres-Ruesta A, Renia L, Ng LF. Type I interferon shapes the quantity and quality of the anti-Zika virus antibody response. Clin Transl Immunology 2020; 9:e1126. [PMID: 32346479 PMCID: PMC7184064 DOI: 10.1002/cti2.1126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/16/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
Objectives Zika virus (ZIKV) is a mosquito-borne flavivirus that re-emerged in 2015. The association between ZIKV and neurological complications initiated the development of relevant animal models to understand the mechanisms underlying ZIKV-induced pathologies. Transient inhibition of the type I interferon (IFN) pathway through the use of an IFNAR1-blocking antibody, MAR1-5A3, could efficiently permit active virus replication in immunocompetent animals. Type I IFN signalling is involved in the regulation of humoral responses, and thus, it is crucial to investigate the potential effects of type I IFN blockade towards B-cell responses. Methods In this study, comparative analysis was conducted using serum samples collected from ZIKV-infected wild-type (WT) animals either administered with or without MAR1-5A3. Results Serological assays revealed a more robust ZIKV-specific IgG response and subtype switching upon inhibition of type I IFN due to the abundance of antigen availability. This observation was corroborated by an increase in germinal centres, plasma cells and germinal centre B cells. Interestingly, although both groups of animals recognised different B-cell linear epitopes in the E and NS1 regions, there was no difference in neutralising capacity. Further characterisation of these epitopes in the E protein revealed a detrimental role of antibodies that were generated in the absence of type I IFN. Conclusion This study highlights the role of type I IFN in shaping the anti-ZIKV antibody response to generate beneficial antibodies and will help guide development of better vaccine candidates triggering efficient neutralising antibodies and avoiding detrimental ones.
Collapse
Affiliation(s)
- Cheryl Yi-Pin Lee
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,NUS Graduate School for Integrative Sciences and Engineering National University of Singapore Singapore
| | - Guillaume Carissimo
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Zheyuan Chen
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,School of Medicine Dentistry & Biomedical Sciences Queen's University Belfast Belfast UK
| | - Fok-Moon Lum
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Farhana Abu Bakar
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,School of Biological Sciences Nanyang Technological University Singapore Singapore
| | - Ravisankar Rajarethinam
- Institute of Molecular and Cell Biology Agency of Science, Technology and Research (ASTAR) Singapore
| | - Teck-Hui Teo
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Present address: Institut Pasteur Unite de Pathogenie Microbienne Moleculaire Paris France
| | - Anthony Torres-Ruesta
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Department of Biochemistry Yong Loo Lin School of Medicine National University of Singapore Singapore
| | - Laurent Renia
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore
| | - Lisa Fp Ng
- Singapore Immunology Network Agency for Science, Technology and Research (ASTAR) Singapore.,Department of Biochemistry Yong Loo Lin School of Medicine National University of Singapore Singapore.,Institute of Infection and Global Health University of Liverpool Liverpool UK
| |
Collapse
|
24
|
Chen X, Li C, Lin W, Li T, Li X, Bai X, Wulin S, Zhang Q, Li S, Liu M, Liu JH, Zhang Y. A Novel Neutralizing Antibody Targeting a Unique Cross-Reactive Epitope on the hi Loop of Domain II of the Envelope Protein Protects Mice against Duck Tembusu Virus. THE JOURNAL OF IMMUNOLOGY 2020; 204:1836-1848. [PMID: 32132180 DOI: 10.4049/jimmunol.1901352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/27/2020] [Indexed: 01/26/2023]
Abstract
The Flavivirus E protein induces protective immunity, and its Abs cause serious problems for serodiagnosis because of the difficulty in differentiating cross-reactive Abs. Moreover, cross-reactive Abs may increase disease severity after secondary Flavivirus infections via Ab-dependent enhancement. Cross-reactive epitopes are therefore critical for understanding serodiagnosis and improving the general knowledge of Flavivirus infections. A minimal epitope, 227GSSAGTWQN235, was identified by a neutralizing mAb 1G2 against duck Tembusu virus (DTMUV), which recognized only monomer E protein under nonreducing conditions. It was unexpectedly found that mutations in the epitope residues G231 or W233 completely abolished reactivity to 1G2 and sera from mice infected with Japanese encephalitis virus, West Nile virus, and Zika virus. An immunofluorescence assay confirmed that mAb 1G2 could cross-react with the E proteins from Japanese encephalitis virus, West Nile virus, and Zika virus. Protein and virus modeling revealed that the epitope was surface accessible in the mature virus and located in the hi loop of domain II. The neutralization of DTMUV by 1G2 played a clear therapeutic role in mouse models. The passive transfer of 1G2 resulted in 100% survival, reduced weight loss, and the complete clearance of DTMUV from the blood of BALB/c mice. Our findings document, for the first time to our knowledge, that mAb 1G2 targets the cross-reactive epitope on the hi loop of domain II in the E protein and might be of potential therapeutic value in treating DTMUV infection and improve the understanding of the issues related to serodiagnosis.
Collapse
Affiliation(s)
- Xueming Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Chenxi Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Weiwei Lin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Tongtong Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Xiaojun Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Xiaofei Bai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Shaozhou Wulin
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Qingshan Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Shuang Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Ming Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| | - Jyung-Hurng Liu
- Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung City 40227, Taiwan
| | - Yun Zhang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute of the Chinese Academy of Agricultural Sciences, Harbin 150069, China; and
| |
Collapse
|
25
|
Sinigaglia A, Peta E, Riccetti S, Barzon L. New avenues for therapeutic discovery against West Nile virus. Expert Opin Drug Discov 2020; 15:333-348. [DOI: 10.1080/17460441.2020.1714586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Elektra Peta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Silvia Riccetti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
26
|
Gil L, Martín A, Lazo L. Wanted Dead or Alive: A Correlate of Protection Against Dengue Virus. Front Immunol 2020; 10:2946. [PMID: 31921194 PMCID: PMC6927490 DOI: 10.3389/fimmu.2019.02946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 12/02/2019] [Indexed: 12/02/2022] Open
Affiliation(s)
- Lázaro Gil
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | | | - Laura Lazo
- Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
27
|
Dobler G, Kaier K, Hehn P, Böhmer MM, Kreusch TM, Borde JP. Tick-borne encephalitis virus vaccination breakthrough infections in Germany: a retrospective analysis from 2001 to 2018. Clin Microbiol Infect 2019; 26:1090.e7-1090.e13. [PMID: 31843655 DOI: 10.1016/j.cmi.2019.12.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/24/2019] [Accepted: 12/02/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES There are few data available regarding the clinical course of tick-borne encephalitis virus (TBEV) vaccination breakthrough infections. The published studies suggest that vaccination breakthrough infections may have a more severe course than native TBEV infection in unvaccinated individuals-potentially due to antibody-dependent enhancement. Here we report a large analysis of vaccination breakthrough infections. METHODS This retrospective analysis was based on a national surveillance dataset spanning the years 2001-2018. Variables reflecting disease severity, such as 'CNS symptoms', 'myelitis', 'fatal outcome' and 'hospitalization' were analysed as well as general epidemiological variables. Cases were categorized as 'unvaccinated' or 'ever vaccinated', the latter category including cases with at least one dose of a TBEV vaccine. RESULTS A total of 6073 notified TBEV infection cases were included in our analysis. Sufficient data on vaccination status were available for 95.1% of patients (5777/6073); of these, 5298 presented with a native infection. A total of (334/5777) cases developed an infection despite having been vaccinated at least once. Comparing unvaccinated patients with those with at least one vaccination, we find an odds ratio (OR) 2.73, (95% confidence interval (CI) 0.79-9.50) regarding the variable fatal outcome that did not reach statistical significance. Analysing the clinical variables 'CNS symptoms' and 'myelitis', there is no difference between these groups (OR 0.86, 95% CI 0.68-1.08; and OR 1.30, 95% CI 0.74-2.27 respectively). Patients who were vaccinated and had an assumed protection at symptom onset (n = 100) had a higher risk for the development of myelitic symptoms (OR 2.21, 95% CI 1.01-4.86]) than unvaccinated patients. CONCLUSION Our findings could neither verify that vaccination breakthrough infections might cause a more severe disease than native infections nor prove a clear antibody-dependent enhancement phenomenon. It remains unclear whether the increased myelitis risk in a subgroup of vaccinated patients is a true effect or confounded.
Collapse
Affiliation(s)
- G Dobler
- Bundeswehr Institute of Microbiology, German National Reference Laboratory for TBEV, Munich, Germany
| | - K Kaier
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Centre - University of Freiburg, Freiburg, Germany
| | - P Hehn
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Centre - University of Freiburg, Freiburg, Germany
| | - M M Böhmer
- Bavarian Health and Food Safety Authority, Department of Infectious Disease Epidemiology & Taskforce Infectiology/Airport, Oberschleissheim, Germany
| | - T M Kreusch
- Robert Koch Institute, Department of Infectious Disease Epidemiology, Immunization Unit, Berlin, Germany
| | - J P Borde
- Division of Infectious Diseases, Department of Medicine II, University of Freiburg Medical Centre and Faculty of Medicine, Freiburg, Germany; Praxis Dr. J. Borde/Gesundheitszentrum Oberkirch, Oberkirch, Germany.
| |
Collapse
|
28
|
Blight J, Alves E, Reyes-Sandoval A. Considering Genomic and Immunological Correlates of Protection for a Dengue Intervention. Vaccines (Basel) 2019; 7:E203. [PMID: 31816907 PMCID: PMC6963661 DOI: 10.3390/vaccines7040203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 01/18/2023] Open
Abstract
Over three billion are at risk of dengue infection with more than 100 million a year presenting with symptoms that can lead to deadly haemorrhagic disease. There are however no treatments available and the only licensed vaccine shows limited efficacy and is able to enhance the disease in some cases. These failures have mainly been due to the complex pathology and lack of understanding of the correlates of protection for dengue virus (DENV) infection. With increasing data suggesting both a protective and detrimental effect for antibodies and CD8 T-cells whilst having complex environmental dynamics. This review discusses the roles of genomic and immunological aspects of DENV infection, providing both a historical interpretation and fresh discussion on how this information can be used for the next generation of dengue interventions.
Collapse
Affiliation(s)
- Joshua Blight
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK; (J.B.); (E.A.)
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Eduardo Alves
- Department of Life Sciences, Imperial College London, Sir Alexander Fleming Building, Exhibition Road, South Kensington, London SW7 2AZ, UK; (J.B.); (E.A.)
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, The Henry Wellcome Building for Molecular Physiology, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
29
|
Flavivirus infection—A review of immunopathogenesis, immunological response, and immunodiagnosis. Virus Res 2019; 274:197770. [DOI: 10.1016/j.virusres.2019.197770] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022]
|
30
|
Gao F, Lin X, He L, Wang R, Wang H, Shi X, Zhang F, Yin C, Zhang L, Zhu J, Yu L. Development of a Potent and Protective Germline-Like Antibody Lineage Against Zika Virus in a Convalescent Human. Front Immunol 2019; 10:2424. [PMID: 31708914 PMCID: PMC6821881 DOI: 10.3389/fimmu.2019.02424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) specific neutralizing antibodies hold great promise for antibody-based interventions and vaccine design against ZIKV infection. However, their development in infected patients remains unclear. Here, we applied next-generation sequencing (NGS) to probe the dynamic development of a potent and protective ZIKV E DIII-specific antibody ZK2B10 isolated from a ZIKV convalescent individual. The unbiased repertoire analysis showed dramatic changes in the usage of antibody variable region germline genes. However, lineage tracing of ZK2B10 revealed limited somatic hypermutation and transient expansion during the 12 months following the onset of symptoms. The NGS-derived, germline-like ZK2B10 somatic variants neutralized ZIKV potently and protected mice from lethal challenge of ZIKV without detectable cross-reactivity with Dengue virus (DENV). Site-directed mutagenesis identified two residues within the λ chain, N31 and S91, that are essential to the functional maturation of ZK2B10. The repertoire and lineage features unveiled here will help elucidate the developmental process and protective potential of E DIII-directed antibodies against ZIKV infection.
Collapse
Affiliation(s)
- Fei Gao
- Department of Basic Medical Sciences, Comprehensive AIDS Research Center, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohe Lin
- Department of Integrative Structural and Computational Biology, Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Linling He
- Department of Integrative Structural and Computational Biology, Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Ruoke Wang
- Department of Basic Medical Sciences, Comprehensive AIDS Research Center, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Han Wang
- Department of Basic Medical Sciences, Comprehensive AIDS Research Center, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Xuanling Shi
- Department of Basic Medical Sciences, Comprehensive AIDS Research Center, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Fuchun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Chibiao Yin
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Linqi Zhang
- Department of Basic Medical Sciences, Comprehensive AIDS Research Center, Beijing Advanced Innovation Center for Structural Biology, School of Medicine, Tsinghua University, Beijing, China
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, United States
| | - Lei Yu
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
31
|
Yellow Fever: Integrating Current Knowledge with Technological Innovations to Identify Strategies for Controlling a Re-Emerging Virus. Viruses 2019; 11:v11100960. [PMID: 31627415 PMCID: PMC6832525 DOI: 10.3390/v11100960] [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: 08/30/2019] [Revised: 09/30/2019] [Accepted: 10/11/2019] [Indexed: 01/17/2023] Open
Abstract
Yellow fever virus (YFV) represents a re-emerging zoonotic pathogen, transmitted by mosquito vectors to humans from primate reservoirs. Sporadic outbreaks of YFV occur in endemic tropical regions, causing a viral hemorrhagic fever (VHF) associated with high mortality rates. Despite a highly effective vaccine, no antiviral treatments currently exist. Therefore, YFV represents a neglected tropical disease and is chronically understudied, with many aspects of YFV biology incompletely defined including host range, host–virus interactions and correlates of host immunity and pathogenicity. In this article, we review the current state of YFV research, focusing on the viral lifecycle, host responses to infection, species tropism and the success and associated limitations of the YFV-17D vaccine. In addition, we highlight the current lack of available treatments and use publicly available sequence and structural data to assess global patterns of YFV sequence diversity and identify potential drug targets. Finally, we discuss how technological advances, including real-time epidemiological monitoring of outbreaks using next-generation sequencing and CRISPR/Cas9 modification of vector species, could be utilized in future battles against this re-emerging pathogen which continues to cause devastating disease.
Collapse
|
32
|
A Single Injection of Human Neutralizing Antibody Protects against Zika Virus Infection and Microcephaly in Developing Mouse Embryos. Cell Rep 2019; 23:1424-1434. [PMID: 29719255 PMCID: PMC7104101 DOI: 10.1016/j.celrep.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/27/2018] [Accepted: 03/30/2018] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted flavivirus that is generally benign in humans. However, an emergent strain of ZIKV has become widespread, causing severe pre- and post-natal neurological defects. There is now an urgent need for prophylactic and therapeutic agents. To address this, we investigated six human monoclonal antibodies with ZIKV epitope specificity and neutralizing activity in mouse models of ZIKV infection and microcephaly. A single intraperitoneal injection of these antibodies conveyed distinct levels of adult and in utero protection from ZIKV infection, which closely mirrored their respective in vitro neutralizing activities. One antibody, ZK2B10, showed the most potent neutralization activity, completely protected uninfected mice, and markedly reduced tissue pathology in infected mice. Thus, ZK2B10 is a promising candidate for the development of antibody-based interventions and informs the rational design of ZIKV vaccine. Human antibodies against ZIKV are tested in mouse models In vitro neutralizing activity correlates with in vivo protection The most potent antibody, ZK2B10, provides protection against infection ZK2B10 markedly delays mortality
Collapse
|
33
|
West Nile Virus and Usutu Virus Co-Circulation in Europe: Epidemiology and Implications. Microorganisms 2019; 7:microorganisms7070184. [PMID: 31248051 PMCID: PMC6680635 DOI: 10.3390/microorganisms7070184] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/20/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023] Open
Abstract
West Nile virus (WNV) and Usutu virus (USUV) are neurotropic mosquito-borne flaviviruses that may infect humans. Although WNV is much more widespread and plays a much larger role in human health, the two viruses are characterized by similar envelope antigens, clinical manifestations, and present overlapping in terms of geographic range of transmission, host, and vector species. This review highlights some of the most relevant aspects of WNV and USUV human infections in Europe, and the possible implications of their co-circulation.
Collapse
|
34
|
Pre-clinical evaluation of a quadrivalent HCV VLP vaccine in pigs following microneedle delivery. Sci Rep 2019; 9:9251. [PMID: 31239471 PMCID: PMC6592879 DOI: 10.1038/s41598-019-45461-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/29/2019] [Indexed: 02/07/2023] Open
Abstract
The introduction of directly acting antiviral agents (DAAs) has produced significant improvements in the ability to cure chronic hepatitis C infection. However, with over 2% of the world’s population infected with HCV, complications arising from the development of cirrhosis of the liver, chronic hepatitis C infection remains the leading indication for liver transplantation. Several modelling studies have indicated that DAAs alone will not be sufficient to eliminate HCV, but if combined with an effective vaccine this regimen would provide a significant advance towards achieving this critical World Health Organisation goal. We have previously generated a genotype 1a, 1b, 2a, 3a HCV virus like particle (VLP) quadrivalent vaccine. The HCV VLPs contain the core and envelope proteins (E1 and E2) of HCV and the vaccine has been shown to produce broad humoral and T cell immune responses following vaccination of mice. In this report we further advanced this work by investigating vaccine responses in a large animal model. We demonstrate that intradermal microneedle vaccination of pigs with our quadrivalent HCV VLP based vaccine produces long-lived multi-genotype specific and neutralizing antibody (NAb) responses together with strong T cell and granzyme B responses and normal Th1 and Th2 cytokine responses. These responses were achieved without the addition of adjuvant. Our study demonstrates that our vaccine is able to produce broad immune responses in a large animal that, next to primates, is the closest animal model to humans. Our results are important as they show that the vaccine can produce robust immune responses in a large animal model before progressing the vaccine to human trials.
Collapse
|
35
|
Yamanaka A, Konishi E. Key Amino Acid Substitution for Infection-Enhancing Activity-Free Designer Dengue Vaccines. iScience 2019; 13:125-137. [PMID: 30826727 PMCID: PMC6402262 DOI: 10.1016/j.isci.2019.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Dengue is a globally important disease caused by four serotypes of dengue virus. Dengue vaccine development has been hampered by antigenic cross-reactivity among serotypes, which potentially causes antibody-dependent enhancement of infection and disease severity. Here we found that a single amino acid substitution in the envelope protein at position 87 from aspartic acid to asparagine or at position 107 from leucine to phenylalanine is critical for suppressing the induction of infection-enhancing antibody in a mouse model. The site and type of amino acid substitution were determined via neutralization escape using an enhancing-activity-only monoclonal antibody that was engineered to reveal neutralizing activity. Mutated dengue type 1 DNA vaccines containing either or both amino acid substitutions induced neutralizing antibodies devoid of enhancing activity against all serotypes. The effect of substitution was further demonstrated using other serotypes and a tetravalent formulation. This finding may contribute to the development of infection-enhancing-activity-free dengue vaccines. Amino acids at E87 or E107 are critical for dengue-enhancing antibody induction Neutralization escape is useful for identifying the key types or sites of amino acids Each substitution can be applied to antigens of all four dengue serotypes A modified tetravalent DNA vaccine suppresses enhancing antibody induction in mice
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
36
|
Isa DM, Chin SP, Chong WL, Zain SM, Rahman NA, Lee VS. Dynamics and binding interactions of peptide inhibitors of dengue virus entry. J Biol Phys 2019; 45:63-76. [PMID: 30680580 DOI: 10.1007/s10867-018-9515-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
In this study, we investigate the binding interactions of two synthetic antiviral peptides (DET2 and DET4) on type II dengue virus (DENV2) envelope protein domain III. These two antiviral peptides are designed based on the domain III of the DENV2 envelope protein, which has shown significant inhibition activity in previous studies and can be potentially modified further to be active against all dengue strains. Molecular docking was performed using AutoDock Vina and the best-ranked peptide-domain III complex was further explored using molecular dynamics simulations. Molecular mechanics-Poisson-Boltzmann surface area (MM-PBSA) was used to calculate the relative binding free energies and to locate the key residues of peptide-protein interactions. The predicted binding affinity correlated well with the previous experimental studies. DET4 outperformed DET2 and is oriented within the binding site through favorable vdW and electrostatic interactions. Pairwise residue decomposition analysis has revealed several key residues that contribute to the binding of these peptides. Residues in DET2 interact relatively lesser with the domain III compared to DET4. Dynamic cross-correlation analysis showed that both the DET2 and DET4 trigger different dynamic patterns on the domain III. Correlated motions were seen between the residue pairs of DET4 and the binding site while binding of DET2 results in anti-correlated motion on the binding site. This work showcases the use of computational study in elucidating and explaining the experiment observation on an atomic level.
Collapse
Affiliation(s)
- Diyana Mohd Isa
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sek Peng Chin
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Wei Lim Chong
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sharifuddin M Zain
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Noorsaadah Abd Rahman
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Vannajan Sanghiran Lee
- Department of Chemistry, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
37
|
Rojas JM, Rodríguez-Martín D, Avia M, Martín V, Sevilla N. Peste des Petits Ruminants Virus Fusion and Hemagglutinin Proteins Trigger Antibody-Dependent Cell-Mediated Cytotoxicity in Infected Cells. Front Immunol 2019; 9:3172. [PMID: 30693004 PMCID: PMC6339941 DOI: 10.3389/fimmu.2018.03172] [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: 08/31/2018] [Accepted: 12/24/2018] [Indexed: 11/25/2022] Open
Abstract
The adaptive immune system utilizes multiple effector mechanisms to clear viral infections. Among those antibody-dependent cell-mediated cytotoxicity (ADCC) can help recognize and clear virus-infected cells. In the present work we evaluated ADCC contribution to immunity in two economically important viral diseases that affect ruminants: bluetongue and peste des petits ruminants. Immune sera obtained from sheep experimentally infected with bluetongue virus (BTV) serotype 8 or peste des petits ruminant virus (PPRV) IC'89 were used for this study. PPRV immune sera could bind to the surface of PPRV-infected ovine B cells while BTV immune sera was unable to bind to the surface of BTV-infected sheep cells but could recognize intracellular BTV antigens. BTV and PPRV immune serum ADCC potency was established using an ovine autologous cytotoxicity assay that employed an NK cell-enriched fraction as effector cells and a virus-infected B cell-enriched fraction as target cells. In this system, immune sera triggered ADCC against PPRV-infected cells, but not against BTV-infected cells. PPRV immune sera could recognize PPRV fusion and hemagglutinin proteins on the surface of transfected cells, and enhanced lysis of these cells in ADCC assays. This indicated that these viral antigens are natural ADCC targets during PPRV infection. The present work describes a novel effector immune mechanism against PPRV in the natural host that could contribute to virus clearance highlighting the importance of studying protective immune mechanisms to improve current vaccines by invoking all effector arms of immunity.
Collapse
Affiliation(s)
- José M Rojas
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA), Madrid, Spain
| | - Daniel Rodríguez-Martín
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA), Madrid, Spain
| | - Miguel Avia
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA), Madrid, Spain
| | - Verónica Martín
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA), Madrid, Spain
| | - Noemí Sevilla
- Centro de Investigación en Sanidad Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (CISA-INIA), Madrid, Spain
| |
Collapse
|
38
|
Sinigaglia L, Gracias S, Décembre E, Fritz M, Bruni D, Smith N, Herbeuval JP, Martin A, Dreux M, Tangy F, Jouvenet N. Immature particles and capsid-free viral RNA produced by Yellow fever virus-infected cells stimulate plasmacytoid dendritic cells to secrete interferons. Sci Rep 2018; 8:10889. [PMID: 30022130 PMCID: PMC6052170 DOI: 10.1038/s41598-018-29235-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 07/09/2018] [Indexed: 12/18/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are specialized in the production of interferons (IFNs) in response to viral infections. The Flaviviridae family comprises enveloped RNA viruses such as Hepatitis C virus (HCV) and Dengue virus (DENV). Cell-free flaviviridae virions poorly stimulate pDCs to produce IFN. By contrast, cells infected with HCV and DENV potently stimulate pDCs via short-range delivery of viral RNAs, which are either packaged within immature virions or secreted exosomes. We report that cells infected with Yellow fever virus (YFV), the prototypical flavivirus, stimulated pDCs to produce IFNs in a TLR7- and cell contact- dependent manner. Such stimulation was unaffected by the presence of YFV neutralizing antibodies. As reported for DENV, cells producing immature YFV particles were more potent at stimulating pDCs than cells releasing mature virions. Additionally, cells replicating a release-deficient YFV mutant or a YFV subgenomic RNA lacking structural protein-coding sequences participated in pDC stimulation. Thus, viral RNAs produced by YFV-infected cells reach pDCs via at least two mechanisms: within immature particles and as capsid-free RNAs. Our work highlights the ability of pDCs to respond to a variety of viral RNA-laden carriers generated from infected cells.
Collapse
Affiliation(s)
- Laura Sinigaglia
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Ségolène Gracias
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Elodie Décembre
- CIRI, Inserm U1111, CNRS UMR5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Matthieu Fritz
- Molecular Genetics of RNA Viruses Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Daniela Bruni
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Nikaïa Smith
- Chemistry & Biology, Modeling & Immunology for Therapy, UMR8601 CNRS, Université Paris Descartes, Paris, France
| | - Jean-Philippe Herbeuval
- Chemistry & Biology, Modeling & Immunology for Therapy, UMR8601 CNRS, Université Paris Descartes, Paris, France
| | - Annette Martin
- Molecular Genetics of RNA Viruses Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Marlène Dreux
- CIRI, Inserm U1111, CNRS UMR5308, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Frédéric Tangy
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Institut Pasteur, Paris, France
| | - Nolwenn Jouvenet
- Viral Genomics and Vaccination Unit, UMR3569 CNRS, Institut Pasteur, Paris, France.
| |
Collapse
|
39
|
Dengue virus serotype distribution based on serological evidence in pediatric urban population in Indonesia. PLoS Negl Trop Dis 2018; 12:e0006616. [PMID: 29953438 PMCID: PMC6040755 DOI: 10.1371/journal.pntd.0006616] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/11/2018] [Accepted: 06/16/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Dengue is a febrile illness transmitted by mosquitoes, causing disease across the tropical and sub-tropical world. Antibody prevalence data and serotype distributions describe population-level risk and inform public health decision-making. METHODOLOGY/PRINCIPAL FINDINGS In this cross-sectional study we used data from a pediatric dengue seroprevalence study to describe historical dengue serotype circulation, according to age and geographic location. A sub-sample of 780 dengue IgG-positive sera, collected from 30 sites across urban Indonesia in 2014, were tested by the plaque reduction neutralization test (PRNT) to measure the prevalence and concentration of serotype-specific neutralizing antibodies according to subject age and geography. PRNT results were obtained from 776 subjects with mean age of 9.6 years. 765 (98.6%) neutralized one or more dengue serotype at a threshold of >10 (1/dil). Multitypic profiles were observed in 50.9% of the samples; a proportion which increased to 63.1% in subjects aged 15-18 years. Amongst monotypic samples, the highest proportion was reactive against DENV-2, followed by DENV-1, and DENV-3, with some variation across the country. DENV-4 was the least common serotype. The highest anti-dengue antibody titers were recorded against DENV-2, and increased with age to a geometric mean of 516.5 [1/dil] in the oldest age group. CONCLUSIONS/SIGNIFICANCE We found that all four dengue serotypes have been widely circulating in most of urban Indonesia, and more than half of children had already been exposed to >1 dengue serotype, demonstrating intense transmission often associated with more severe clinical episodes. These data will help inform policymakers and highlight the importance of dengue surveillance, prevention and control.
Collapse
|
40
|
Fernandez E, Kose N, Edeling MA, Adhikari J, Sapparapu G, Lazarte SM, Nelson CA, Govero J, Gross ML, Fremont DH, Crowe JE, Diamond MS. Mouse and Human Monoclonal Antibodies Protect against Infection by Multiple Genotypes of Japanese Encephalitis Virus. mBio 2018; 9:e00008-18. [PMID: 29487230 PMCID: PMC5829823 DOI: 10.1128/mbio.00008-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 01/26/2018] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis virus (JEV) remains a leading cause of viral encephalitis worldwide. Although JEV-specific antibodies have been described, an assessment of their ability to neutralize multiple genotypes of JEV has been limited. Here, we describe the development of a panel of mouse and human neutralizing monoclonal antibodies (MAbs) that inhibit infection in cell culture of four different JEV genotypes tested. Mechanism-of-action studies showed that many of these MAbs inhibited infection at a postattachment step, including blockade of virus fusion. Mapping studies using site-directed mutagenesis and hydrogen-deuterium exchange with mass spectrometry revealed that the lateral ridge on domain III of the envelope protein was a primary recognition epitope for our panel of strongly neutralizing MAbs. Therapeutic studies in mice demonstrated protection against lethality caused by genotype I and III strains when MAbs were administered as a single dose even 5 days after infection. This information may inform the development of vaccines and therapeutic antibodies as emerging strains and genotypic shifts become more prevalent.IMPORTANCE Although Japanese encephalitis virus (JEV) is a vaccine-preventable cause of viral encephalitis, the inactivated and live attenuated platforms available are derived from strains belonging to a single genotype (GIII) due to its historical prevalence in areas of JEV epidemics. Related to this, studies with vaccines and antibodies have focused on assessing the in vitro and in vivo protective responses to homologous or heterologous GIII strains. An epidemiological shift in JEV genotype distribution warrants the induction of broadly neutralizing antibody responses that inhibit infection of multiple JEV genotypes. Here, we generated a panel of mouse and human neutralizing monoclonal antibodies and evaluated their inhibitory activity, epitope location, and capacity for protection against multiple JEV genotypes in mice.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/immunology
- Chlorocebus aethiops
- Disease Models, Animal
- Encephalitis Virus, Japanese/classification
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Epitopes/immunology
- Genotype
- Humans
- Mice
- Models, Biological
- Treatment Outcome
- Vero Cells
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Estefania Fernandez
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Nurgun Kose
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Melissa A Edeling
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jagat Adhikari
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Gopal Sapparapu
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Susana M Lazarte
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Christopher A Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Jennifer Govero
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - James E Crowe
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, USA
| |
Collapse
|
41
|
Rey FA, Stiasny K, Vaney MC, Dellarole M, Heinz FX. The bright and the dark side of human antibody responses to flaviviruses: lessons for vaccine design. EMBO Rep 2018; 19:206-224. [PMID: 29282215 PMCID: PMC5797954 DOI: 10.15252/embr.201745302] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 01/07/2023] Open
Abstract
Zika and dengue viruses belong to the Flavivirus genus, a close group of antigenically related viruses that cause significant arthropod-transmitted diseases throughout the globe. Although infection by a given flavivirus is thought to confer lifelong protection, some of the patient's antibodies cross-react with other flaviviruses without cross-neutralizing. The original antigenic sin phenomenon may amplify such antibodies upon subsequent heterologous flavivirus infection, potentially aggravating disease by antibody-dependent enhancement (ADE). The most striking example is provided by the four different dengue viruses, where infection by one serotype appears to predispose to more severe disease upon infection by a second one. A similar effect was postulated for sequential infections with Zika and dengue viruses. In this review, we analyze the molecular determinants of the dual antibody response to flavivirus infection or vaccination in humans. We highlight the role of conserved partially cryptic epitopes giving rise to cross-reacting and poorly neutralizing, ADE-prone antibodies. We end by proposing a strategy for developing an epitope-focused vaccine approach to avoid eliciting undesirable antibodies while focusing the immune system on producing protective antibodies only.
Collapse
Affiliation(s)
- Félix A Rey
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Marie-Christine Vaney
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Mariano Dellarole
- Structural Virology Unit, Virology Department, Institut Pasteur, Paris, France
- CNRS UMR 3569, Paris, France
| | - Franz X Heinz
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
42
|
Abstract
Flaviviruses such as dengue (DENV), yellow fever (YFV), West Nile (WNV), and Zika (ZIKV) are human pathogens of global significance. In particular, DENV causes the most prevalent mosquito-borne viral diseases in humans, and ZIKV emerged from obscurity into the spotlight in 2016 as the etiologic agent of congenital Zika syndrome. Owing to the recent emergence of ZIKV as a global pandemic threat, the roles of the immune system during ZIKV infections are as yet unclear. In contrast, decades of DENV research implicate a dual role for the immune system in protection against and pathogenesis of DENV infection. As DENV and ZIKV are closely related, knowledge based on DENV studies has been used to prioritize investigation of ZIKV immunity and pathogenesis, and to accelerate ZIKV diagnostic, therapeutic, and vaccine design. This review discusses the following topics related to innate and adaptive immune responses to DENV and ZIKV: the interferon system as the key mechanism of host defense and viral target for immune evasion, antibody-mediated protection versus antibody-dependent enhancement, and T cell-mediated protection versus original T cell antigenic sin. Understanding the mechanisms that regulate the balance between immune-mediated protection and pathogenesis during DENV and ZIKV infections is critical toward development of safe and effective DENV and ZIKV therapeutics and vaccines.
Collapse
Affiliation(s)
- Annie Elong Ngono
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California 92037, USA;
| |
Collapse
|
43
|
Development of Antibody Therapeutics against Flaviviruses. Int J Mol Sci 2017; 19:ijms19010054. [PMID: 29295568 PMCID: PMC5796004 DOI: 10.3390/ijms19010054] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/28/2022] Open
Abstract
Recent outbreaks of Zika virus (ZIKV) highlight the urgent need to develop efficacious interventions against flaviviruses, many of which cause devastating epidemics around the world. Monoclonal antibodies (mAb) have been at the forefront of treatment for cancer and a wide array of other diseases due to their specificity and potency. While mammalian cell-produced mAbs have shown promise as therapeutic candidates against several flaviviruses, their eventual approval for human application still faces several challenges including their potential risk of predisposing treated patients to more severe secondary infection by a heterologous flavivirus through antibody-dependent enhancement (ADE). The high cost associated with mAb production in mammalian cell cultures also poses a challenge for the feasible application of these drugs to the developing world where the majority of flavivirus infection occurs. Here, we review the current therapeutic mAb candidates against various flaviviruses including West Nile (WNV), Dengue virus (DENV), and ZIKV. The progress of using plants for developing safer and more economical mAb therapeutics against flaviviruses is discussed within the context of their expression, characterization, downstream processing, neutralization, and in vivo efficacy. The progress of using plant glycoengineering to address ADE, the major impediment of flavivirus therapeutic development, is highlighted. These advancements suggest that plant-based systems are excellent alternatives for addressing the remaining challenges of mAb therapeutic development against flavivirus and may facilitate the eventual commercialization of these drug candidates.
Collapse
|
44
|
Abstract
The emergence of Zika virus (ZIKV) as a major public health threat has focused research on understanding virus biology and developing a suite of strategies for disease intervention. Recent advances in cryoelectron microscopy have accelerated structure-function studies of flaviviruses and of ZIKV in particular. Structures of the mature and immature ZIKV have demonstrated its similarity with other known flaviviruses such as dengue and West Nile viruses. However, ZIKV's unique pathobiology demands an explanation of how its structure, although similar to its flavivirus relatives, is sufficiently unique to address questions of receptor specificity, transmission, and antigenicity. Progress in defining the immunodominant epitopes and how neutralizing antibodies bind to them will provide great insight as vaccines progress through clinical trials. Identification of host receptors will substantially illuminate the interesting ZIKV tropism and provide insights into pathogenesis. Although the answers to all of these questions are not yet available, rapid progress in combining structural biology with other techniques is revealing the similarities and the differences in virion structure and function between ZIKV and related flaviviruses.
Collapse
Affiliation(s)
- Devika Sirohi
- Department of Biological Sciences, Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana
| | - Richard J Kuhn
- Department of Biological Sciences, Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana
| |
Collapse
|
45
|
Tottey S, Shoji Y, Jones RM, Chichester JA, Green BJ, Musiychuk K, Si H, Manceva SD, Rhee A, Shamloul M, Norikane J, Guimarães RC, Caride E, Silva ANMR, Simões M, Neves PCC, Marchevsky R, Freire MS, Streatfield SJ, Yusibov V. Plant-Produced Subunit Vaccine Candidates against Yellow Fever Induce Virus Neutralizing Antibodies and Confer Protection against Viral Challenge in Animal Models. Am J Trop Med Hyg 2017; 98:420-431. [PMID: 29231157 DOI: 10.4269/ajtmh.16-0293] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Yellow fever (YF) is a viral disease transmitted by mosquitoes and endemic mostly in South America and Africa with 20-50% fatality. All current licensed YF vaccines, including YF-Vax® (Sanofi-Pasteur, Lyon, France) and 17DD-YFV (Bio-Manguinhos, Rio de Janeiro, Brazil), are based on live attenuated virus produced in hens' eggs and have been widely used. The YF vaccines are considered safe and highly effective. However, a recent increase in demand for YF vaccines and reports of rare cases of YF vaccine-associated fatal adverse events have provoked interest in developing a safer YF vaccine that can be easily scaled up to meet this increased global demand. To this point, we have engineered the YF virus envelope protein (YFE) and transiently expressed it in Nicotiana benthamiana as a stand-alone protein (YFE) or as fusion to the bacterial enzyme lichenase (YFE-LicKM). Immunogenicity and challenge studies in mice demonstrated that both YFE and YFE-LicKM elicited virus neutralizing (VN) antibodies and protected over 70% of mice from lethal challenge infection. Furthermore, these two YFE-based vaccine candidates induced VN antibody responses with high serum avidity in nonhuman primates and these VN antibody responses were further enhanced after challenge infection with the 17DD strain of YF virus. These results demonstrate partial protective efficacy in mice of YFE-based subunit vaccines expressed in N. benthamiana. However, their efficacy is inferior to that of the live attenuated 17DD vaccine, indicating that formulation development, such as incorporating a more suitable adjuvant, may be required for product development.
Collapse
Affiliation(s)
- Stephen Tottey
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Yoko Shoji
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - R Mark Jones
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Brian J Green
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Huaxin Si
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | | | - Amy Rhee
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Moneim Shamloul
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Joey Norikane
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| | - Rosane C Guimarães
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Elena Caride
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Andrea N M R Silva
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marisol Simões
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Patricia C C Neves
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Renato Marchevsky
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | - Marcos S Freire
- Instituto de Tecnologia em Imunobiológicos, Bio-Manguinhos, Fiocruz, Rio de Janeiro, Brazil
| | | | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware
| |
Collapse
|
46
|
Wen J, Elong Ngono A, Regla-Nava JA, Kim K, Gorman MJ, Diamond MS, Shresta S. Dengue virus-reactive CD8 + T cells mediate cross-protection against subsequent Zika virus challenge. Nat Commun 2017; 8:1459. [PMID: 29129917 PMCID: PMC5682281 DOI: 10.1038/s41467-017-01669-z] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/05/2017] [Indexed: 01/28/2023] Open
Abstract
Zika virus (ZIKV) and dengue virus (DENV) are antigenically related flaviviruses that share cross-reactivity in antibody and T cell responses, and co-circulate in increasing numbers of countries. Whether pre-existing DENV immunity can cross-protect or enhance ZIKV infection during sequential infection of the same host is unknown. Here, we show that DENV-immune Ifnar1−/− or wild-type C57BL/6 mice infected with ZIKV have cross-reactive immunity to subsequent ZIKV infection and pathogenesis. Adoptive transfer and cell depletion studies demonstrate that DENV-immune CD8+ T cells predominantly mediate cross-protective responses to ZIKV. In contrast, passive transfer studies suggest that DENV-immune serum does not protect against ZIKV infection. Thus, CD8+ T cell immunity generated during primary DENV infection can confer protection against secondary ZIKV infection in mice. Further optimization of current DENV vaccines for T cell responses might confer cross-protection and prevent antibody-mediated enhancement of ZIKV infection. Dengue virus-specific antibody and CD8+ T cells that cross-react with Zika virus have been described. Here, the authors establish a functionally protective role for cross-reactive dengue virus-specific CD8+ T cells during challenge with Zika virus.
Collapse
Affiliation(s)
- Jinsheng Wen
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, 92037, USA.,Institute of Arboviruses, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Annie Elong Ngono
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, 92037, USA
| | - Jose Angel Regla-Nava
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, 92037, USA
| | - Matthew J Gorman
- Department of Medicine, Molecular Microbiology, Pathology and Immunology, The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Michael S Diamond
- Department of Medicine, Molecular Microbiology, Pathology and Immunology, The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, 92037, USA. .,Institute of Arboviruses, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China. .,Department of Medicine, School of Medicine, University of California, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
47
|
Sarathy VV, Pitcher TJ, Gromowski GD, Roehrig JT, Barrett ADT. A DENV-2-type-specific monoclonal antibody binds to the DENV-complex-reactive antigenic site on envelope protein domain 3. J Gen Virol 2017. [PMID: 28631593 DOI: 10.1099/jgv.0.000785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The Dengue virus (DENV) envelope (E) protein is the major component of the viral surface and is structurally subdivided into three domains, ED1, ED2 and ED3. ED3 elicits potent neutralizing antibodies and contains two major antigenic sites: the DENV-type-specific and DENV-complex-reactive antigenic sites. Each site is composed of a limited subset of residues that are required for monoclonal antibody (mAb) binding. Here we show that DENV-2-type-specific mAb 9A3D-8 utilizes the functionally critical residues K307, V308, K310, I312, P332, L387, L389 and N390 for ED3 binding. Surprisingly, this DENV-type-specific epitope is predicted to overlap with the ED3 DENV-complex-reactive antigenic site on the viral surface. Further, this unique binding site enables mAb 9A3D-8 to neutralize virus infectivity at relatively low occupancy of virions compared to other ED3 mAbs identified to date. Together, the data in this study indicate that this is a new DENV-2-type-specific antigenic site on ED3.
Collapse
Affiliation(s)
- Vanessa V Sarathy
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Trevor J Pitcher
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Present address: The Binding Site, Inc., San Diego, CA, USA
| | - Gregory D Gromowski
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.,Present address: Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - John T Roehrig
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, 80523, USA
| | - Alan D T Barrett
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston TX 77555, USA.,Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
48
|
Yamanaka A, Konishi E. Dengue-Immune Humans Have Higher Levels of Complement-Independent Enhancing Antibody than Complement-Dependent Neutralizing Antibody. Jpn J Infect Dis 2017; 70:579-581. [PMID: 28367878 DOI: 10.7883/yoken.jjid.2016.379] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Dengue is the most important arboviral disease worldwide. We previously reported that most inhabitants of dengue-endemic countries who are naturally immune to the disease have infection-enhancing antibodies whose in vitro activity does not decrease in the presence of complement (complement-independent enhancing antibodies, or CiEAb). Here, we compared levels of CiEAb and complement-dependent neutralizing antibodies (CdNAb) in dengue-immune humans. A typical antibody dose-response pattern obtained in our assay system to measure the balance between neutralizing and enhancing antibodies showed both neutralizing and enhancing activities depending on serum dilution factor. The addition of complement to the assay system increased the activity of neutralizing antibodies at lower dilutions, indicating the presence of CdNAb. In contrast, similar dose-response curves were obtained with and without complement at higher dilutions, indicating higher levels of CiEAb than CdNAb. For experimental support for the higher CiEAb levels, a cocktail of mouse monoclonal antibodies against dengue virus type 1 was prepared. The antibody dose-response curves obtained in this assay, with or without complement, were similar to those obtained with human serum samples when a high proportion of D1-V-3H12 (an antibody exhibiting only enhancing activity and thus a model for CiEAb) was used in the cocktail. This study revealed higher-level induction of CiEAb than CdNAb in humans naturally infected with dengue viruses.
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University.,BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University.,Center for Infectious Diseases, Kobe University Graduate School of Medicine
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University.,BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University.,Center for Infectious Diseases, Kobe University Graduate School of Medicine
| |
Collapse
|
49
|
Yamanaka A, Moi ML, Takasaki T, Kurane I, Konishi E. Neutralizing and enhancing antibody responses to five genotypes of dengue virus type 1 (DENV-1) in DENV-1 patients. J Gen Virol 2017; 98:166-172. [PMID: 27911254 DOI: 10.1099/jgv.0.000669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue virus (DENV) has four distinct serotypes, DENV-1-4, with four to six genotypes in each serotype. The World Health Organization recommends tetravalent formulations including one genotype of each serotype as safe and effective dengue vaccines. Here, we investigated the impact of genotype on the neutralizing antibody responses to DENV-1 in humans. Convalescent sera collected from patients with primary infection of DENV-1 were examined for neutralizing antibody against single-round infectious particles of the five DENV-1 genotypes (GI-GV). In both GI- and GIV-infected patients, their neutralizing antibody titres against the five genotypes were similar, differing ≤4-fold from the homogenotypic responses. The enhancing activities against the five genotypes were also similar in these sera. Thus, the genotype strains of DENV-1 showed no significant antigenic differences in these patients, suggesting that GI- or GIV-derived vaccine antigens should induce equivalent levels of neutralizing antibodies against all DENV-1 genotypes.
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand.,BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Meng Ling Moi
- Present address: Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan.,Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Tomohiko Takasaki
- Present address: Kanagawa Prefectural Institute of Public Health, Kanagawa, Japan.,Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Ichiro Kurane
- National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.,BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand
| |
Collapse
|
50
|
The Antigenic Structure of Zika Virus and Its Relation to Other Flaviviruses: Implications for Infection and Immunoprophylaxis. Microbiol Mol Biol Rev 2017; 81:81/1/e00055-16. [PMID: 28179396 DOI: 10.1128/mmbr.00055-16] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Zika virus was discovered ∼70 years ago in Uganda and maintained a low profile as a human disease agent in Africa and Asia. Only recently has it caused explosive outbreaks in previously unaffected regions, first in Oceania and then in the Americas since 2015. Of special concern is the newly identified link between congenital malformations (especially microcephaly) and Zika virus infections during pregnancy. At present, it is unclear whether Zika virus changed its pathogenicity or whether the huge number of infections allowed the recognition of a previously cryptic pathogenic property. The purpose of this review is to discuss recent data on the molecular antigenic structure of Zika virus in the context of antibody-mediated neutralization and antibody-dependent enhancement (ADE) of infection, a phenomenon that has been implicated in the development of severe disease caused by the related dengue viruses. Emphasis is given to epitopes of antibodies that potently neutralize Zika virus and also to epitopes that provide antigenic links to other important human-pathogenic flaviviruses such as dengue, yellow fever, West Nile, Japanese encephalitis, and tick-borne encephalitis viruses. The antigenic cross talk between Zika and dengue viruses appears to be of special importance, since they cocirculate in many regions of endemicity and sequential infections are likely to occur frequently. New insights into the molecular antigenic structure of Zika virus and flaviviruses in general have provided the foundation for great progress made in developing Zika virus vaccines and antibodies for passive immunization.
Collapse
|