1
|
Crampon E, Covernton E, Vaney MC, Dellarole M, Sommer S, Sharma A, Haouz A, England P, Lepault J, Duquerroy S, Rey FA, Barba-Spaeth G. New insight into flavivirus maturation from structure/function studies of the yellow fever virus envelope protein complex. mBio 2023; 14:e0070623. [PMID: 37607061 PMCID: PMC10653854 DOI: 10.1128/mbio.00706-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/16/2023] [Indexed: 08/24/2023] Open
Abstract
IMPORTANCE All enveloped viruses enter cells by fusing their envelope with a target cell membrane while avoiding premature fusion with membranes of the producer cell-the latter being particularly important for viruses that bud at internal membranes. Flaviviruses bud in the endoplasmic reticulum, are transported through the TGN to reach the external milieu, and enter other cells via receptor-mediated endocytosis. The trigger for membrane fusion is the acidic environment of early endosomes, which has a similar pH to the TGN of the producer cell. The viral particles therefore become activated to react to mildly acidic pH only after their release into the neutral pH extracellular environment. Our study shows that for yellow fever virus (YFV), the mechanism of activation involves actively knocking out the fusion brake (protein pr) through a localized conformational change of the envelope protein upon exposure to the neutral pH external environment. Our study has important implications for understanding the molecular mechanism of flavivirus fusion activation in general and points to an alternative way of interfering with this process as an antiviral treatment.
Collapse
Affiliation(s)
- E. Crampon
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - E. Covernton
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - M. C. Vaney
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - M. Dellarole
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - S. Sommer
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - A. Sharma
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - A. Haouz
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Plateforme de Cristallographie-C2RT, Paris, France
| | - P. England
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Plateforme de Biophysique Moléculaire-C2RT, Paris, France
| | - J. Lepault
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - S. Duquerroy
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
- Université Paris-Saclay, Faculté des Sciences, Orsay, France
| | - F. A. Rey
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| | - G. Barba-Spaeth
- Institut Pasteur, Université Paris Cité, CNRS UMR 3569, Unité de Virologie Structurale, Paris, France
| |
Collapse
|
2
|
Ishida K, Yagi H, Kato Y, Morita E. N-linked glycosylation of flavivirus E protein contributes to viral particle formation. PLoS Pathog 2023; 19:e1011681. [PMID: 37819933 PMCID: PMC10593244 DOI: 10.1371/journal.ppat.1011681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/23/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023] Open
Abstract
In the case of the Japanese encephalitis virus (JEV), the envelope protein (E), a major component of viral particles, contains a highly conserved N-linked glycosylation site (E: N154). Glycosylation of the E protein is thought to play an important role in the ability of the virus to attach to target cells during transmission; however, its role in viral particle formation and release remains poorly understood. In this study, we investigated the role of N-glycosylation of flaviviral structural proteins in viral particle formation and secretion by introducing mutations in viral structural proteins or cellular factors involved in glycoprotein transport and processing. The number of secreted subviral particles (SVPs) was significantly reduced in N154A, a glycosylation-null mutant, but increased in D67N, a mutant containing additional glycosylation sites, indicating that the amount of E glycosylation regulates the release of SVPs. SVP secretion was reduced in cells deficient in galactose, sialic acid, and N-acetylglucosamine modifications in the Golgi apparatus; however, these reductions were not significant, suggesting that glycosylation mainly plays a role in pre-Golgi transport. Fluorescent labeling of SVPs using a split green fluorescent protein (GFP) system and time-lapse imaging by retention using selective hooks (RUSH) system revealed that the glycosylation-deficient mutant was arrested before endoplasmic reticulum (ER)- Golgi transport. However, the absence of ERGIC-53 and ERGIC-L, ER-Golgi transport cargo receptors that recognize sugar chains on cargo proteins, does not impair SVP secretion. In contrast, the solubility of the N154A mutant of E or the N15A/T17A mutant of prM in cells was markedly lower than that of the wild type, and proteasome-mediated rapid degradation of these mutants was observed, indicating the significance of glycosylation of both prM and E in proper protein folding and assembly of viral particles in the ER.
Collapse
Affiliation(s)
- Kotaro Ishida
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Japan
- Division of Biomolecular Function, Bioresources Science, United Graduate School of Agricultural Sciences, Iwate University, Morioka, Japan
| | - Hirokazu Yagi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eiji Morita
- Department of Biochemistry and Molecular Biology, Faculty of Agriculture and Life Science, Hirosaki University, Japan
- Division of Biomolecular Function, Bioresources Science, United Graduate School of Agricultural Sciences, Iwate University, Morioka, Japan
| |
Collapse
|
3
|
Neurovirulence, viscerotropism and immunogenicity of live attenuated yellow fever 17D vaccine virus in non-human primates. Vaccine 2023; 41:836-843. [PMID: 36564277 DOI: 10.1016/j.vaccine.2022.12.029] [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: 12/11/2021] [Revised: 11/24/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Yellow fever vaccine associated neurovirulence and viscerotropism have been reported by various countries. In this study, the neurovirulence, viscerotropism and immunogenicity of yellow fever vaccine seed lots (master and working) and final product manufactured at Serum Institute of India (SII) were evaluated in cynomolgus monkeys. WHO reference virus 168-73 and Stamaril™ as a control vaccine was used for comparison. Neurovirulence and viscerotropism scores of the seed lots and final product were lower than Stamaril™. The SII seed virus and vaccine complies to the WHO requirement for neurovirulence, viscerotropism and immunogenicity, when tested in comparison to WHO reference seed virus 168/73. All challenged animals showed 100 % seroconversion as early as day 14 and neutralizing antibody titers were sustainable at day 30 in all animals.
Collapse
|
4
|
Gianchecchi E, Cianchi V, Torelli A, Montomoli E. Yellow Fever: Origin, Epidemiology, Preventive Strategies and Future Prospects. Vaccines (Basel) 2022; 10:372. [PMID: 35335004 PMCID: PMC8955180 DOI: 10.3390/vaccines10030372] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 12/10/2022] Open
Abstract
Yellow fever (YF) virus still represents a major threat in low resource countries in both South America and Africa despite the presence of an effective vaccine. YF outbreaks are not only due to insufficient vaccine coverage for insufficient vaccine supply, but also to the increase in people without history of vaccination living in endemic areas. Globalization, continuous population growth, urbanization associated with inadequate public health infrastructure, and climate changes constitute important promoting factors for the spread of this virus to tropical and subtropical areas in mosquito-infested regions capable of spreading the disease. In the present review, we focus on the origin of the virus and its transmission, representing two debated topics throughout the nineteenth century, going deeply into the history of YF vaccines until the development of the vaccine still used nowadays. Besides surveillance, we highlight the urgent need of routine immunization and vaccination campaigns associated to diverse and innovative mosquito control technologies in endemic areas for YF virus in order to minimize the risk of new YF outbreaks and the global burden of YF in the future.
Collapse
Affiliation(s)
- Elena Gianchecchi
- VisMederi Srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy; (V.C.); (E.M.)
| | - Virginia Cianchi
- VisMederi Srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy; (V.C.); (E.M.)
| | | | - Emanuele Montomoli
- VisMederi Srl, Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy; (V.C.); (E.M.)
- Department of Molecular and Developmental Medicine, University of Siena, Via Aldo 3, 53100 Siena, Italy
| |
Collapse
|
5
|
Haslwanter D, Lasso G, Wec AZ, Furtado ND, Raphael LMS, Tse AL, Sun Y, Stransky S, Pedreño-Lopez N, Correia CA, Bornholdt ZA, Sakharkar M, Avelino-Silva VI, Moyer CL, Watkins DI, Kallas EG, Sidoli S, Walker LM, Bonaldo MC, Chandran K. Genotype-specific features reduce the susceptibility of South American yellow fever virus strains to vaccine-induced antibodies. Cell Host Microbe 2022; 30:248-259.e6. [PMID: 34998466 PMCID: PMC10067022 DOI: 10.1016/j.chom.2021.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022]
Abstract
The resurgence of yellow fever in South America has prompted vaccination against the etiologic agent, yellow fever virus (YFV). Current vaccines are based on a live-attenuated YF-17D virus derived from a virulent African isolate. The capacity of these vaccines to induce neutralizing antibodies against the vaccine strain is used as a surrogate for protection. However, the sensitivity of genetically distinct South American strains to vaccine-induced antibodies is unknown. We show that antiviral potency of the polyclonal antibody response in vaccinees is attenuated against an emergent Brazilian strain. This reduction was attributable to amino acid changes at two sites in central domain II of the glycoprotein E, including multiple changes at the domain I-domain II hinge, which are unique to and shared among most South American YFV strains. Our findings call for a reevaluation of current approaches to YFV immunological surveillance in South America and suggest approaches for updating vaccines.
Collapse
Affiliation(s)
- Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | | | - Nathália Dias Furtado
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil
| | - Lidiane Menezes Souza Raphael
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil
| | - Alexandra L Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Yan Sun
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Núria Pedreño-Lopez
- Department of Pathology, The George Washington University, Washington, DC 20037, USA
| | - Carolina Argondizo Correia
- Laboratório de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | | | | | - Vivian I Avelino-Silva
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | | | - David I Watkins
- Department of Pathology, The George Washington University, Washington, DC 20037, USA
| | - Esper G Kallas
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Laura M Walker
- Adimab, LLC, Lebanon, NH 03766, USA; Adagio Therapeutics Inc., Waltham, MA 02451, USA
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA.
| |
Collapse
|
6
|
Genome Characterization of Yellow Fever Virus Wild-Type Strain Asibi, Parent to Live-Attenuated 17D Vaccine, from Three Different Sources. Viruses 2021; 13:v13071383. [PMID: 34372589 PMCID: PMC8310116 DOI: 10.3390/v13071383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 11/16/2022] Open
Abstract
The yellow fever virus vaccine, 17D, was derived through the serial passage of the wild-type (WT) strain Asibi virus in mouse and chicken tissue. Since its derivation, the mechanism of attenuation of 17D virus has been investigated using three 17D substrains and WT Asibi virus. Although all three substrains of 17D have been sequenced, only one isolate of Asibi has been examined genetically and all interpretation of attenuation is based on this one isolate. Here, we sequenced the genome of Asibi virus from three different laboratories and show that the WT strain is genetically homogenous at the amino acids that distinguish Asibi from 17D vaccine virus.
Collapse
|
7
|
Cordero-Rivera CD, De Jesús-González LA, Osuna-Ramos JF, Palacios-Rápalo SN, Farfan-Morales CN, Reyes-Ruiz JM, Del Ángel RM. The importance of viral and cellular factors on flavivirus entry. Curr Opin Virol 2021; 49:164-175. [PMID: 34171540 DOI: 10.1016/j.coviro.2021.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The flavivirus are emerging and re-emerging arthropod-borne pathogens responsible for significant mortality and morbidity worldwide. The genus comprises more than 70 viruses, and despite genomic and structural similarities, infections by different flaviviruses result in different clinical presentations. In the absence of a safe and effective vaccine against these infections, the search for new strategies to inhibit viral infection is necessary. The life cycle of arboviruses begins with the entry process composed of multiple steps: attachment, internalization, endosomal escape and capsid uncoating. This mini-review describes factors and mechanisms involved in the viral entry as events required to take over the cellular machinery and host factors and cellular pathways commonly used by flaviviruses as possible approaches for developing broad-spectrum antiviral drugs.
Collapse
Affiliation(s)
- Carlos Daniel Cordero-Rivera
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - Luis Adrián De Jesús-González
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - Juan Fidel Osuna-Ramos
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - Selvin Noé Palacios-Rápalo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - Carlos Noe Farfan-Morales
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - José Manuel Reyes-Ruiz
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico
| | - Rosa María Del Ángel
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Intituto Politécnico Nacional (CINVESTAV-IPN), Ciudad de México 07320, Mexico.
| |
Collapse
|
8
|
Serrão de Andrade AA, Soares AER, Paula de Almeida LG, Ciapina LP, Pestana CP, Aquino CL, Medeiros MA, Ribeiro de Vasconcelos AT. Testing the genomic stability of the Brazilian yellow fever vaccine strain using next-generation sequencing data. Interface Focus 2021; 11:20200063. [PMID: 34123353 PMCID: PMC8193464 DOI: 10.1098/rsfs.2020.0063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2021] [Indexed: 01/06/2023] Open
Abstract
The live attenuated yellow fever (YF) vaccine was developed in the 1930s. Currently, the 17D and 17DD attenuated substrains are used for vaccine production. The 17D strain is used for vaccine production by several countries, while the 17DD strain is used exclusively in Brazil. The cell passages carried out through the seed-lot system of vaccine production influence the presence of quasispecies causing changes in the stability and immunogenicity of attenuated genotypes by increasing attenuation or virulence. Using next-generation sequencing, we carried out genomic characterization and genetic diversity analysis between vaccine lots of the Brazilian YF vaccine, produced by BioManguinhos–Fiocruz, and used during 11 years of vaccination in Brazil. We present 20 assembled and annotated genomes from the Brazilian 17DD vaccine strain, eight single nucleotide polymorphisms and the quasispecies spectrum reconstruction for the 17DD vaccine, through a pipeline here introduced. The V2IDA pipeline provided a relationship between low genetic diversity, maintained through the seed lot system, and the confirmation of genetic stability of lots of the Brazilian vaccine against YF. Our study sets precedents for use of V2IDA in genetic diversity analysis and in silico stability investigation of attenuated viral vaccines, facilitating genetic surveillance during the vaccine production process.
Collapse
Affiliation(s)
- Amanda Araújo Serrão de Andrade
- National Laboratory for Scientific Computing, Bioinformatics Laboratory (LABINFO), Avenida Getúlio Vargas, 333, Quitandinha 25651-075, Petrópolis, Rio de Janeiro, Brazil
| | - André E R Soares
- National Laboratory for Scientific Computing, Bioinformatics Laboratory (LABINFO), Avenida Getúlio Vargas, 333, Quitandinha 25651-075, Petrópolis, Rio de Janeiro, Brazil
| | - Luiz Gonzaga Paula de Almeida
- National Laboratory for Scientific Computing, Bioinformatics Laboratory (LABINFO), Avenida Getúlio Vargas, 333, Quitandinha 25651-075, Petrópolis, Rio de Janeiro, Brazil
| | - Luciane Prioli Ciapina
- National Laboratory for Scientific Computing, Bioinformatics Laboratory (LABINFO), Avenida Getúlio Vargas, 333, Quitandinha 25651-075, Petrópolis, Rio de Janeiro, Brazil
| | - Cristiane Pinheiro Pestana
- Fiocruz, Bio-Manguinhos, Recombinant Technology Laboratory (LATER), Brazilian Ministry of Health, Rio de Janeiro, Brazil
| | - Carolina Lessa Aquino
- Fiocruz, Bio-Manguinhos, Recombinant Technology Laboratory (LATER), Brazilian Ministry of Health, Rio de Janeiro, Brazil
| | - Marco Alberto Medeiros
- Fiocruz, Bio-Manguinhos, Recombinant Technology Laboratory (LATER), Brazilian Ministry of Health, Rio de Janeiro, Brazil
| | - Ana Tereza Ribeiro de Vasconcelos
- National Laboratory for Scientific Computing, Bioinformatics Laboratory (LABINFO), Avenida Getúlio Vargas, 333, Quitandinha 25651-075, Petrópolis, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Flavivirus Envelope Protein Glycosylation: Impacts on Viral Infection and Pathogenesis. J Virol 2020; 94:JVI.00104-20. [PMID: 32161171 DOI: 10.1128/jvi.00104-20] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Flaviviruses encode one, two, or no N-linked glycosylation sites on their envelope proteins. Glycosylation can impact virus interactions with cell surface attachment factors and also may impact virion stability and virus replication. Envelope protein glycosylation has been identified as a virulence determinant for multiple flaviviruses, but the mechanisms by which glycosylation mediates pathogenesis remain unclear. In this Gem, we summarize current knowledge on flavivirus envelope protein glycosylation and its impact on viral infection and pathogenesis.
Collapse
|
10
|
Fontes-Garfias CR, Shan C, Luo H, Muruato AE, Medeiros DBA, Mays E, Xie X, Zou J, Roundy CM, Wakamiya M, Rossi SL, Wang T, Weaver SC, Shi PY. Functional Analysis of Glycosylation of Zika Virus Envelope Protein. Cell Rep 2018; 21:1180-1190. [PMID: 29091758 DOI: 10.1016/j.celrep.2017.10.016] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/29/2017] [Accepted: 10/04/2017] [Indexed: 02/08/2023] Open
Abstract
Zika virus (ZIKV) infection causes devastating congenital abnormities and Guillain-Barré syndrome. The ZIKV envelope (E) protein is responsible for viral entry and represents a major determinant for viral pathogenesis. Like other flaviviruses, the ZIKV E protein is glycosylated at amino acid N154. To study the function of E glycosylation, we generated a recombinant N154Q ZIKV that lacks the E glycosylation and analyzed the mutant virus in mammalian and mosquito hosts. In mouse models, the mutant was attenuated, as evidenced by lower viremia, decreased weight loss, and no mortality; however, knockout of E glycosylation did not significantly affect neurovirulence. Mice immunized with the mutant virus developed a robust neutralizing antibody response and were completely protected from wild-type ZIKV challenge. In mosquitoes, the mutant virus exhibited diminished oral infectivity for the Aedes aegypti vector. Collectively, the results demonstrate that E glycosylation is critical for ZIKV infection of mammalian and mosquito hosts.
Collapse
Affiliation(s)
- Camila R Fontes-Garfias
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Chao Shan
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Huanle Luo
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Antonio E Muruato
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Daniele B A Medeiros
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ministry of Health, Ananindeua, Pará State, Brazil
| | - Elizabeth Mays
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Xuping Xie
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jing Zou
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Christopher M Roundy
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA
| | - Maki Wakamiya
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology and Center for Biodefense & Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott C Weaver
- Institute for Human Infections & Immunity, University of Texas Medical Branch, Galveston, TX, USA; Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
11
|
Gong D, Zhang TH, Zhao D, Du Y, Chapa TJ, Shi Y, Wang L, Contreras D, Zeng G, Shi PY, Wu TT, Arumugaswami V, Sun R. High-Throughput Fitness Profiling of Zika Virus E Protein Reveals Different Roles for Glycosylation during Infection of Mammalian and Mosquito Cells. iScience 2018; 1:97-111. [PMID: 30227960 PMCID: PMC6135943 DOI: 10.1016/j.isci.2018.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 01/26/2018] [Accepted: 02/12/2018] [Indexed: 12/29/2022] Open
Abstract
Zika virus (ZIKV) infection causes Guillain-Barré syndrome and severe birth defects. ZIKV envelope (E) protein is the major viral protein involved in cell receptor binding and entry and is therefore considered one of the major determinants in ZIKV pathogenesis. Here we report a gene-wide mapping of functional residues of ZIKV E protein using a mutant library, with changes covering every nucleotide position. By comparing the replication fitness of every viral mutant between mosquito and human cells, we identified that mutations affecting glycosylation display the most divergence. By characterizing individual mutants, we show that ablation of glycosylation selectively benefits ZIKV infection of mosquito cells by enhancing cell entry, whereas it either has little impact on ZIKV infection on certain human cells or leads to decreased infection through the entry factor DC-SIGN. In conclusion, we define the roles of individual residues of ZIKV envelope protein, which contribute to ZIKV replication fitness in human and mosquito cells.
Collapse
Affiliation(s)
- Danyang Gong
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Tian-Hao Zhang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Dawei Zhao
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yushen Du
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Travis J Chapa
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yuan Shi
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Laurie Wang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | - Deisy Contreras
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gang Zeng
- Department of Urology, University of California, Los Angeles, CA 90095, USA
| | - Pei-Yong Shi
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | | | - Ren Sun
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
12
|
Mossenta M, Marchese S, Poggianella M, Slon Campos J, Burrone O. Role of N-glycosylation on Zika virus E protein secretion, viral assembly and infectivity. Biochem Biophys Res Commun 2017; 492:579-586. [DOI: 10.1016/j.bbrc.2017.01.022] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/05/2017] [Indexed: 01/03/2023]
|
13
|
Roby JA, Setoh YX, Hall RA, Khromykh AA. Post-translational regulation and modifications of flavivirus structural proteins. J Gen Virol 2015; 96:1551-69. [PMID: 25711963 DOI: 10.1099/vir.0.000097] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Flaviviruses are a group of single-stranded, positive-sense RNA viruses that generally circulate between arthropod vectors and susceptible vertebrate hosts, producing significant human and veterinary disease burdens. Intensive research efforts have broadened our scientific understanding of the replication cycles of these viruses and have revealed several elegant and tightly co-ordinated post-translational modifications that regulate the activity of viral proteins. The three structural proteins in particular - capsid (C), pre-membrane (prM) and envelope (E) - are subjected to strict regulatory modifications as they progress from translation through virus particle assembly and egress. The timing of proteolytic cleavage events at the C-prM junction directly influences the degree of genomic RNA packaging into nascent virions. Proteolytic maturation of prM by host furin during Golgi transit facilitates rearrangement of the E proteins at the virion surface, exposing the fusion loop and thus increasing particle infectivity. Specific interactions between the prM and E proteins are also important for particle assembly, as prM acts as a chaperone, facilitating correct conformational folding of E. It is only once prM/E heterodimers form that these proteins can be secreted efficiently. The addition of branched glycans to the prM and E proteins during virion transit also plays a key role in modulating the rate of secretion, pH sensitivity and infectivity of flavivirus particles. The insights gained from research into post-translational regulation of structural proteins are beginning to be applied in the rational design of improved flavivirus vaccine candidates and make attractive targets for the development of novel therapeutics.
Collapse
Affiliation(s)
- Justin A Roby
- 1Australian Infectious Diseases Research Centre, The University of Queensland, Australia 2School of Chemistry and Molecular Biosciences, The University of Queensland, Australia
| | - Yin Xiang Setoh
- 1Australian Infectious Diseases Research Centre, The University of Queensland, Australia 2School of Chemistry and Molecular Biosciences, The University of Queensland, Australia
| | - Roy A Hall
- 1Australian Infectious Diseases Research Centre, The University of Queensland, Australia 2School of Chemistry and Molecular Biosciences, The University of Queensland, Australia
| | - Alexander A Khromykh
- 1Australian Infectious Diseases Research Centre, The University of Queensland, Australia 2School of Chemistry and Molecular Biosciences, The University of Queensland, Australia
| |
Collapse
|
14
|
Beasley DWC, McAuley AJ, Bente DA. Yellow fever virus: genetic and phenotypic diversity and implications for detection, prevention and therapy. Antiviral Res 2014; 115:48-70. [PMID: 25545072 DOI: 10.1016/j.antiviral.2014.12.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 11/28/2022]
Abstract
Yellow fever virus (YFV) is the prototypical hemorrhagic fever virus, yet our understanding of its phenotypic diversity and any molecular basis for observed differences in disease severity and epidemiology is lacking, when compared to other arthropod-borne and haemorrhagic fever viruses. This is, in part, due to the availability of safe and effective vaccines resulting in basic YFV research taking a back seat to those viruses for which no effective vaccine occurs. However, regular outbreaks occur in endemic areas, and the spread of the virus to new, previously unaffected, areas is possible. Analysis of isolates from endemic areas reveals a strong geographic association for major genotypes, and recent epidemics have demonstrated the emergence of novel sequence variants. This review aims to outline the current understanding of YFV genetic and phenotypic diversity and its sources, as well as the available animal models for characterizing these differences in vivo. The consequences of genetic diversity for detection and diagnosis of yellow fever and development of new vaccines and therapeutics are discussed.
Collapse
Affiliation(s)
- David W C Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States.
| | - Alexander J McAuley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| | - Dennis A Bente
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Sealy Center for Vaccine Development, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States; Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, United States
| |
Collapse
|
15
|
Yellow fever vaccine: Comparison of the neurovirulence of new 17D-204 Stamaril™ seed lots and RK 168-73 strain. Biologicals 2013; 41:238-46. [DOI: 10.1016/j.biologicals.2013.04.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/17/2013] [Accepted: 04/20/2013] [Indexed: 11/24/2022] Open
|
16
|
Beasley DWC, Morin M, Lamb AR, Hayman E, Watts DM, Lee CK, Trent DW, Monath TP. Adaptation of yellow fever virus 17D to Vero cells is associated with mutations in structural and non-structural protein genes. Virus Res 2013; 176:280-4. [PMID: 23602827 DOI: 10.1016/j.virusres.2013.04.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/04/2013] [Accepted: 04/09/2013] [Indexed: 12/14/2022]
Abstract
Serial passaging of yellow fever virus 17D in Vero cells was employed to derive seed material for a novel inactivated vaccine, XRX-001. Two independent passaging series identified a novel lysine to arginine mutation at amino acid 160 of the envelope protein, a surface-exposed residue in structural domain I. A third passage series resulted in an isoleucine to methionine mutation at residue 113 of the NS4B protein, a central membrane spanning region of the protein which has previously been associated with Vero cell adaptation of other mosquito-borne flaviviruses. These studies confirm that flavivirus adaptation to growth in Vero cells can be mediated by structural or non-structural protein mutations.
Collapse
Affiliation(s)
- David W C Beasley
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
|
18
|
Moudy RM, Payne AF, Dodson BL, Kramer LD. Requirement of glycosylation of West Nile virus envelope protein for infection of, but not spread within, Culex quinquefasciatus mosquito vectors. Am J Trop Med Hyg 2011; 85:374-8. [PMID: 21813861 DOI: 10.4269/ajtmh.2011.10-0697] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Most of sequenced West Nile virus (WNV) genomes encode a single N-linked glycosylation site on their envelope (E) proteins. We previously found that WNV lacking the E protein glycan was severely inhibited in its ability to replicate and spread within two important mosquito vector species, Culex pipiens and Cx. tarsalis. However, recent work with a closely related species, Cx. pipiens pallens, found no association between E protein glycosylation and either replication or dissemination. To examine this finding further, we expanded upon our previous studies to include an additional Culex species, Cx. quinquefasciatus. The non-glycosylated WNV-N154I virus replicated less efficiently in mosquito tissues after intrathoracic inoculation, but there was little difference in replication efficiency in the midgut after peroral infection. Interestingly, although infectivity was inhibited when WNV lacked the E protein glycan, there was little difference in viral spread throughout the mosquito. These data indicate that E protein glycosylation affects WNV-vector interactions in a species-specific manner.
Collapse
Affiliation(s)
- Robin M Moudy
- New York State Department of Health, Slingerlands, NY, USA.
| | | | | | | |
Collapse
|
19
|
Neurovirulence tests of three 17D yellow fever vaccine strains. Biologicals 2011; 39:167-70. [DOI: 10.1016/j.biologicals.2011.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 04/01/2011] [Accepted: 04/03/2011] [Indexed: 11/30/2022] Open
|
20
|
Both E protein glycans adversely affect dengue virus infectivity but are beneficial for virion release. J Virol 2010; 84:5171-80. [PMID: 20219924 DOI: 10.1128/jvi.01900-09] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The E protein of most flaviviruses is modified by Asn-linked glycosylation at residue 153/154 and in the case of the four dengue virus (DENV) serotypes by a second glycan at residue 67. However, the absence of E protein glycosylation among numerous natural isolates of different flaviviruses suggests that the glycan, per se, is not critically important in the virus life cycle. Consistent with this notion, we show that ablation of both glycans from the DENV-2 E protein reduces but does not prevent growth of the variant in mammalian and mosquito cells. We found a pronounced and opposing effect of glycan ablation on two stages of the virus growth cycle: infectivity and release. Loss of either of the two DENV E protein glycans markedly enhanced infectivity of variants for mosquito cells at the expense of efficient virion release. The variants also displayed reduced release in mammalian cells, which was more prominent for viruses lacking the Asn 67-linked glycan than for those lacking the Asn 153-linked glycan, without a marked change in infectivity. Mutations, which compensated for the defect in virus morphogenesis associated with ablation of the Asn 67-linked glycan in mammalian cells but interestingly not in mosquito cells, were identified at the glycosylation acceptor motif and a second site in E protein domain II. The dueling influences of infectivity and release on virus growth affected by the glycans may explain the plasticity in E protein glycosylation among the flaviviruses.
Collapse
|
21
|
Maciel M, Kellathur SN, Chikhlikar P, Dhalia R, Sidney J, Sette A, August TJ, Marques ET. Comprehensive analysis of T cell epitope discovery strategies using 17DD yellow fever virus structural proteins and BALB/c (H2d) mice model. Virology 2008; 378:105-17. [PMID: 18579176 PMCID: PMC2615555 DOI: 10.1016/j.virol.2008.04.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2008] [Revised: 04/07/2008] [Accepted: 04/30/2008] [Indexed: 01/19/2023]
Abstract
Immunomics research uses in silico epitope prediction, as well as in vivo and in vitro approaches. We inoculated BALB/c (H2d) mice with 17DD yellow fever vaccine to investigate the correlations between approaches used for epitope discovery: ELISPOT assays, binding assays, and prediction software. Our results showed a good agreement between ELISPOT and binding assays, which seemed to correlate with the protein immunogenicity. PREDBALB/c prediction software partially agreed with the ELISPOT and binding assay results, but presented low specificity. The use of prediction software to exclude peptides containing no epitopes, followed by high throughput screening of the remaining peptides by ELISPOT, and the use of MHC-biding assays to characterize the MHC restrictions demonstrated to be an efficient strategy. The results allowed the characterization of 2 MHC class I and 17 class II epitopes in the envelope protein of the YF virus in BALB/c (H2d) mice.
Collapse
Affiliation(s)
- Milton Maciel
- Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA
| | - Srinivasan N. Kellathur
- Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA,Johns Hopkins Singapore, Singapore
| | - Pryia Chikhlikar
- Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA
| | - Rafael Dhalia
- Oswaldo Cruz Foundation (FIOCRUZ), Instituto Aggeu Magalhaes, Recife, Brazil
| | | | | | - Thomas J. August
- Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA
| | - Ernesto T.A. Marques
- Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA,Johns Hopkins, School of Medicine, Department of Infectious Diseases, Baltimore, USA,Oswaldo Cruz Foundation (FIOCRUZ), Instituto Aggeu Magalhaes, Recife, Brazil,Corresponding author. Johns Hopkins University, School of Medicine, Pharmacology Department, Baltimore, USA
| |
Collapse
|
22
|
|
23
|
Barban V, Girerd Y, Aguirre M, Gulia S, Pétiard F, Riou P, Barrere B, Lang J. High stability of yellow fever 17D-204 vaccine: A 12-year restrospective analysis of large-scale production. Vaccine 2007; 25:2941-50. [PMID: 16914238 DOI: 10.1016/j.vaccine.2006.06.082] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 05/31/2006] [Accepted: 06/20/2006] [Indexed: 11/22/2022]
Abstract
We have retrospectively analyzed 12 bulk lots of yellow fever vaccine Stamaril, produced between 1990 and 2002 and prepared from the same seed lot that has been in continuous use since 1990. All vaccine batches displayed identical genome sequence. Only four nucleotide substitutions were observed, compared to previously published sequence, with no incidence at amino-acid level. Fine analysis of viral plaque size distribution was used as an additional marker for genetic stability and demonstrated a remarkable homogeneity of the viral population. The total virus load, measured by qRT-PCR, was also homogeneous pointing out reproducibility of the vaccine production process. Mice inoculated intracerebrally with the different bulks exhibited a similar average survival time, and ratio between in vitro potency and mouse LD(50) titers remained constant from batch-to-batch. Taken together, these data demonstrate the genetic stability of the strain at mass production level over a period of 12 years and reinforce the generally admitted idea of the safety of YF17D-based vaccines.
Collapse
Affiliation(s)
- V Barban
- Research Department, Sanofi Pasteur, Marcy-l'Etoile, France.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Mateu GP, Marchevsky RS, Liprandi F, Bonaldo MC, Coutinho ESF, Dieudonné M, Caride E, Jabor AV, Freire MS, Galler R. Construction and biological properties of yellow fever 17D/dengue type 1 recombinant virus. Trans R Soc Trop Med Hyg 2007; 101:289-98. [PMID: 17169387 DOI: 10.1016/j.trstmh.2006.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Revised: 08/23/2006] [Accepted: 08/24/2006] [Indexed: 11/21/2022] Open
Abstract
Dengue virus, a mosquito-borne flavivirus, is one of the most formidable public health threats in tropical and subtropical regions. As yet, there is no licensed vaccine to protect against the disease. A chimeric yellow fever (YF) 17D/dengue (DEN) type 1 virus was constructed by replacing the pre-membrane and envelope genes of YF 17D virus with those from DEN 1 VeMir95 virus, a Venezuelan isolate. The chimeric YF 17D/DEN 1 VeMir95 virus was regenerated from full-length infectious clones stably propagated in Escherichia coli by transfection of Vero cells with in vitro transcribed RNA. The chimeric virus proliferated efficiently in Vero cells ( approximately 6.6 log(10) plaque-forming units/ml). The chimeric virus was not neurovirulent to 3-week-old Swiss Webster mice inoculated by the intracerebral route, in contrast to the YF 17DD vaccine strain that was lethal for 90% of the mice. The YF 17D/DEN 1 virus at Passage 6 was more attenuated for rhesus monkeys than the YF 17DD commercial vaccine after intracerebral inoculation according to the standard neurovirulence test. This virus is a potential candidate to be included in a tetravalent DEN vaccine formulation. The availability of the cloned cDNA allows further structure/function studies on the viral envelope.
Collapse
Affiliation(s)
- G P Mateu
- Fundacão Oswaldo Cruz, Departamento de Bioquimica e Biología Molecular, Avenida Brasil 4365, Manguinhos, 21045-900 Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Yellow fever, a mosquito-borne viral hemorrhagic fever, is one of the most lethal diseases of humankind. The etiologic agent is the prototype member of the genus Flavivirus, family Flaviviridae, a group of small, enveloped, positive-sense, single-strand RNA viruses. Approximately one in seven people who become infected develop a rapidly progressive illness, with hepatitis, renal failure, hemorrhage and cardiovascular shock, with a case fatality rate of 20-50%. Yellow fever occurs in sub-Saharan Africa and tropical South America, where it remains a continuing public health problem of varying magnitude, depending on the level of vaccination coverage in the human population and cyclical, ecologic and climatic factors that influence virus transmission.
Collapse
|
26
|
Barba-Spaeth G, Longman RS, Albert ML, Rice CM. Live attenuated yellow fever 17D infects human DCs and allows for presentation of endogenous and recombinant T cell epitopes. ACTA ACUST UNITED AC 2005; 202:1179-84. [PMID: 16260489 PMCID: PMC2213233 DOI: 10.1084/jem.20051352] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The yellow fever (YF) 17D vaccine is one of the most successful live attenuated vaccines available. A single immunization induces both long-lasting neutralizing antibody and YF-specific T cell responses. Surprisingly, the mechanism for this robust immunity has not been addressed. In light of several recent reports suggesting flavivirus interaction with dendritic cells (DCs), we investigated the mechanism of YF17D interaction with DCs and the importance of this interaction in generating T cell immunity. Our results show that YF17D can infect immature and mature human DCs. Viral entry is Ca2+ dependent, but it is independent of DC-SIGN as well as multiple integrins expressed on the DC surface. Similar to infection of cell lines, YF infection of immature DCs is cytopathic. Although infection itself does not induce DC maturation in vitro, TNF-α–induced maturation protects DCs from YF-induced cytopathogenicity. Furthermore, we show that DCs infected with YF17D or YF17D carrying a recombinant epitope can process and present antigens for CD8+ T cell stimulation. These findings offer insight into the immunologic mechanisms associated with the highly capable YF17D vaccine that may guide effective vaccine design.
Collapse
Affiliation(s)
- Giovanna Barba-Spaeth
- Laboratory of Virology and Infectious Disease, Center for the Study of Hepatitis C, The Rockefeller University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
27
|
Affiliation(s)
- Franz X Heinz
- Institute of Virology, University of Vienna, A-1095 Vienna, Austria
| | | |
Collapse
|
28
|
Goto A, Hayasaka D, Yoshii K, Mizutani T, Kariwa H, Takashima I. A BHK-21 cell culture-adapted tick-borne encephalitis virus mutant is attenuated for neuroinvasiveness. Vaccine 2003; 21:4043-51. [PMID: 12922141 DOI: 10.1016/s0264-410x(03)00269-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We derived the baby hamster kidney (BHK)-21 cell culture-adapted, tick-borne encephalitis (TBE) virus mutant. To reveal the pathogenicity of the TBE virus, we compared the pathogenicity of the mutant (Oshima Cl-1) and parental (Oshima 5-10) virus in mouse model. The neurovirulence of mutant in mice was identical to that of parent. However, the level of neuroinvasiveness was higher for parent than for mutant. The degrees of viremia and virus titers in the spleen were lower in mice that were inoculated subcutaneously (s.c.) with mutant than in mice that received parent. Unlike parent, mutant was rarely detected in the brains of s.c. inoculated mice. Genetic analysis revealed that mutant had single amino acid substitutions in each of the E and NS5 proteins compared with parent. Furthermore, while mutant infection of BHK-21 cells was inhibited by glycosaminoglycans (GAGs), this was not the case for parent. In summary, the BHK-21-cell-adapted mutant virus showed reduced neuroinvasiveness in mice due to low-level induction of viremia. The attenuation process involved a single amino acid change in the E protein, which may have resulted in the rapid clearance of the virus due to its high affinity for negatively charged molecules in vivo.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Viral/analysis
- Antibodies, Viral/biosynthesis
- Brain/pathology
- Brain/virology
- Cell Line
- Cricetinae
- Encephalitis Viruses, Tick-Borne/genetics
- Encephalitis Viruses, Tick-Borne/immunology
- Encephalitis Viruses, Tick-Borne/pathogenicity
- Encephalitis, Tick-Borne/pathology
- Encephalitis, Tick-Borne/prevention & control
- Encephalitis, Tick-Borne/virology
- Gene Products, gag/biosynthesis
- Genome, Viral
- Hemagglutination Tests
- Male
- Mice
- Mice, Inbred ICR
- Molecular Sequence Data
- Mutation/genetics
- Mutation/immunology
- Neutralization Tests
- Spleen/virology
- Vaccines, Attenuated/immunology
- Viral Plaque Assay
- Viral Vaccines/immunology
- Viremia/blood
- Virus Replication
Collapse
Affiliation(s)
- Akiko Goto
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Kita-18 Nishi-9, Kita-ku, Sapporo 060-0818, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
It will be apparent to the reader that there is much to learn about the pathogenesis of YF. The role of specific genes and molecular determinants of neurotropism and viscerotropism has been defined only partially. The availability of infectious clones and a small animal (hamster) model should allow dissection of virulence factors, which can then be tested in the more difficult monkey model. The marked differences between wild-type YF strains should be evaluated by evaluating the relationships between virulence and genome sequence. The role of cytokine dysregulation and endothelial injury in YF will be elucidated as access to patients and of patients to more sophisticated medical care improves. The number of cases of YF in unvaccinated travelers hospitalized after return from the tropics has unfortunately increased, but such cases afford unique opportunities to study the pathogenesis of renal failure, coagulopathy, vascular instability, and shock, as well as new treatment modalities. At the cellular level, there are also important opportunities for research on YF virus-cell receptor interactions, the control of apoptotic cell death, and the predilection for cells of the midzone of the liver lobule. The role of dendritic cells in the early stage of YF infection is deserving of study. Finally, the role of the immune response to infection, particularly cellular immunity, is poorly characterized, and the suggestion that immune clearance may aggravate the condition of the host during the period of intoxication should be evaluated in appropriate animal models.
Collapse
Affiliation(s)
- Thomas P Monath
- Acambis Inc. and Department of Microbiology and Immunology, Harvard School of Public Health, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
30
|
Bonaldo MC, Garratt RC, Caufour PS, Freire MS, Rodrigues MM, Nussenzweig RS, Galler R. Surface expression of an immunodominant malaria protein B cell epitope by yellow fever virus. J Mol Biol 2002; 315:873-85. [PMID: 11812154 DOI: 10.1006/jmbi.2001.5258] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The yellow fever 17D virus (YF17D) has several characteristics that are desirable for the development of new, live attenuated vaccines. We approached its development as a vector for heterologous antigens by studying the expression of a humoral epitope at the surface of the E protein based on the results of modelling its three-dimensional structure. This model indicated that the most promising insertion site is between beta-strands f and g, a site that is exposed at the external surface of the virus. The large deletion of six residues from the fg loop of the E protein from yellow fever virus, compared to tick-born encephalitis virus, leaves space at the dimer interface for a large insertion without creating steric hindrance. We have tested this hypothesis by inserting a model humoral epitope from the circumsporozoite protein of Plasmodium falciparum consisting of triple NANP repeats. Recombinant virus (17D/8) expressing this insertion flanked by two glycine residues at each end, is specifically neutralized by a monoclonal antibody to the model epitope. Furthermore, mouse antibodies raised to the recombinant virus recognize the parasite protein in an ELISA assay. Serial passage analysis confirmed the genetic stability of the insertion made in the viral genome and the resulting 17D/8 virus is significantly more attenuated in mouse neurovirulence tests than the 17DD vaccine. The fg loop belongs to the dimerization domain of the E protein and lies at the interface between monomers. This domain undergoes a low pH transition, which is related to the fusion of the viral envelope to the endosome membrane. It is conceivable that a slower rate of fusion, resulting from the insertion close to the dimer interface, may delay the onset of virus production and thereby lead to a milder infection of the host. This would account for the more attenuated phenotype of the recombinant virus in the mouse model and lower extent of replication in cultured cells. The vectorial capacity of the yellow fever virus is being further explored for the expression and presentation of other epitopes, including those mediating T-cell responses.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, Protozoan/chemistry
- Antigens, Protozoan/genetics
- Antigens, Protozoan/immunology
- Cell Line
- Epitopes, B-Lymphocyte/chemistry
- Epitopes, B-Lymphocyte/genetics
- Epitopes, B-Lymphocyte/immunology
- Gene Expression
- Genome, Viral
- Hydrogen-Ion Concentration
- Immunodominant Epitopes/chemistry
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/immunology
- Malaria/immunology
- Malaria/parasitology
- Malaria Vaccines/administration & dosage
- Malaria Vaccines/adverse effects
- Malaria Vaccines/genetics
- Malaria Vaccines/immunology
- Mice
- Models, Molecular
- Molecular Sequence Data
- Neutralization Tests
- Plasmodium falciparum/chemistry
- Plasmodium falciparum/genetics
- Plasmodium falciparum/immunology
- Protein Structure, Tertiary
- Sequence Alignment
- Serial Passage
- Survival Rate
- Vaccines, Attenuated/adverse effects
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Yellow fever virus/genetics
- Yellow fever virus/isolation & purification
- Yellow fever virus/pathogenicity
- Yellow fever virus/physiology
Collapse
Affiliation(s)
- Myrna C Bonaldo
- Departamento de Bioquímica e Biologia Molecular, Fundação Oswaldo Cruz Instituto Oswaldo Cruz, Rio de Janeiro, RJ 21045-900, Brazil
| | | | | | | | | | | | | |
Collapse
|
31
|
Galler R, Pugachev KV, Santos CL, Ocran SW, Jabor AV, Rodrigues SG, Marchevsky RS, Freire MS, Almeida LF, Cruz AC, Yamamura AM, Rocco IM, da Rosa ES, Souza LT, Vasconcelos PF, Guirakhoo F, Monath TP. Phenotypic and molecular analyses of yellow fever 17DD vaccine viruses associated with serious adverse events in Brazil. Virology 2001; 290:309-19. [PMID: 11883195 DOI: 10.1006/viro.2001.1168] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The yellow fever (YF) 17D virus is one of the most successful vaccines developed to data. Its use has been estimated to be over 400 million doses with an excellent record of safety. In the past 3 years, yellow fever vaccination was intensified in Brazil in response to higher risk of urban outbreaks of the disease. Two fatal adverse events temporally associated with YF vaccination were reported. Both cases had features similar to yellow fever disease, including hepatitis and multiorgan failure. Two different lots of YF 17DD virus vaccine were administered to the affected patients and also to hundreds of thousands of other individuals without any other reported serious adverse events. The lots were prepared from the secondary seed, which has been in continuous use since 1984. Nucleotide sequencing revealed minor variations at some nucleotide positions between the secondary seed lot virus and the virus isolates from patients; these differences were not consistent across the isolates, represented differences in the relative amount of each nucleotide in a heterogeneous position, and did not result in amino acid substitutions. Inoculation of rhesus monkeys with the viruses isolated from the two patients by the intracerebral (ic) or intrahepatic (ih) route caused minimal viremia and no clinical signs of infection or alterations in laboratory markers. Central nervous system histological scores of rhesus monkeys inoculated ic were within the expected range, and there were no histopathological lesions in animals inoculated ih. Altogether, these results demonstrated the genetic stability and attenuated phenotype of the viruses that caused fatal illness in the two patients. Therefore, the fatal adverse events experienced by the vaccinees are related to individual, genetically determined host factors that regulate cellular susceptibility to yellow fever virus. Such increased susceptibility, resulting in clinically overt disease expression, appears to be extremely rare.
Collapse
Affiliation(s)
- R Galler
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21045-900, Rio de Janeiro, RJ, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Caufour PS, Motta MC, Yamamura AM, Vazquez S, Ferreira II, Jabor AV, Bonaldo MC, Freire MS, Galler R. Construction, characterization and immunogenicity of recombinant yellow fever 17D-dengue type 2 viruses. Virus Res 2001; 79:1-14. [PMID: 11551641 DOI: 10.1016/s0168-1702(01)00273-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Chimeric yellow fever (YF)-dengue type 2 (Den 2) viruses were constructed by replacing the premembrane (prM) and envelope (E) genes of YF 17D virus with those from Den 2 virus strains of south-east Asian genotype. Whereas viable chimeric viruses were successfully recovered when the YF 17D C gene and the Den 2 prM gene were fused at the signalase cleavage site, no virus could be rescued from the constructions fused at the viral protease cleavage site. Unlike YF virus that replicated in all the cell lines tested and similar to the Den 2 virus, the recombinant viruses did not replicate in vaccine-production certified CEF and MRC5 cells. Besides, chimeric 17D/Den 2 viruses and their parental viruses reached similar growth titers in Vero and C6/36 cell cultures. Analysis of mouse neurovirulence, performed by intracerebral inoculation, demonstrated that the 17D/Den 2 chimera is more attenuated in this system than the YF 17DD virus. Immunization of mice with this chimera induced a neutralizing antibody response associated with a partial protection against an otherwise lethal dose of mouse neurovirulent Den 2 NGC virus. Overall, these results provide further support for the use of chimeric viruses as an attractive methodology for the development of new live flavivirus vaccines.
Collapse
Affiliation(s)
- P S Caufour
- Fundação Oswaldo Cruz, Instituto Oswaldo Cruz, Departamento de Bioquimica e Biologia Molecular, RJ 21045-900, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mandl CW, Kroschewski H, Allison SL, Kofler R, Holzmann H, Meixner T, Heinz FX. Adaptation of tick-borne encephalitis virus to BHK-21 cells results in the formation of multiple heparan sulfate binding sites in the envelope protein and attenuation in vivo. J Virol 2001; 75:5627-37. [PMID: 11356970 PMCID: PMC114275 DOI: 10.1128/jvi.75.12.5627-5637.2001] [Citation(s) in RCA: 178] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Propagation of the flavivirus tick-borne encephalitis virus in BHK-21 cells selected for mutations within the large surface glycoprotein E that increased the net positive charge of the protein. In the course of 16 independent experiments, 12 different protein E mutation patterns were identified. These were located in all three of the structural domains and distributed over almost the entire upper and lateral surface of protein E. The mutations resulted in the formation of local patches of predominantly positive surface charge. Recombinant viruses carrying some of these mutations in a defined genetic backbone showed heparan sulfate (HS)-dependent phenotypes, resulting in an increased specific infectivity and binding affinity for BHK-21 cells, small plaque formation in porcine kidney cells, and significant attenuation of neuroinvasiveness in adult mice. Our results corroborate the notion that the selection of attenuated HS binding mutants is a common and frequent phenomenon during the propagation of viruses in cell culture and suggest a major role for HS dependence in flavivirus attenuation. Recognition of this principle may be of practical value for designing attenuated flavivirus strains in the future.
Collapse
Affiliation(s)
- C W Mandl
- Institute of Virology, University of Vienna, Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
34
|
van der Most RG, Corver J, Strauss JH. Mutagenesis of the RGD motif in the yellow fever virus 17D envelope protein. Virology 1999; 265:83-95. [PMID: 10603320 DOI: 10.1006/viro.1999.0026] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The envelope protein of yellow fever virus 17D (YFV-17D) contains a solvent-exposed RGD motif, which has led to the suggestion that integrins may function as cellular receptors for YFV-17D. We found that mutating the RGD motif to RGE had no effect on viral titers, whereas changing RGD to TGD, TGE, TAD, TAE, or RGS led to reduced titers. Substitution of RGD by RAD or RAE yielded RNA genomes that replicated in mammalian cells but could not spread to neighboring cells at 37 degrees C. These mutants did spread through the cell monolayer at 30 degrees C (both in mosquito cells and in SW13 cells) and viruses grown at this temperature were capable of infecting mammalian cells at 37 degrees C. These results strongly suggest that RGD-mediated integrin binding does not play a major role in YFV-17D entry, since the RGD to RAD mutation, as well as many or all of the other mutations studied, should disrupt all RGD-dependent integrin binding. However, the RGD to RAD or RAE mutations (as well as TAD and TAE) severely destabilized the envelope protein at 37 degrees C, providing an explanation for the observed phenotype. Implications of these findings are discussed in light of the fact that mutations that alter tropism or virulence in different flaviviruses are often found within the loop containing the RGD motif.
Collapse
Affiliation(s)
- R G van der Most
- Division of Biology 156-29, California Institute of Technology, Pasadena, California, 91125, USA
| | | | | |
Collapse
|
35
|
Guirakhoo F, Zhang ZX, Chambers TJ, Delagrave S, Arroyo J, Barrett AD, Monath TP. Immunogenicity, genetic stability, and protective efficacy of a recombinant, chimeric yellow fever-Japanese encephalitis virus (ChimeriVax-JE) as a live, attenuated vaccine candidate against Japanese encephalitis. Virology 1999; 257:363-72. [PMID: 10329547 DOI: 10.1006/viro.1999.9695] [Citation(s) in RCA: 126] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Yellow fever (YF) 17D vaccine virus, having a 60-year history of safe and effective use, is an ideal vector to deliver heterologous genes from other medically important flaviviruses. A chimeric YF/Japanese encephalitis (JE) virus (ChimeriVax-JE virus) was constructed by insertion of the premembrane and envelope (prME) genes of an attenuated human vaccine strain (SA14-14-2) of Japanese encephalitis (JE) virus between core and nonstructural (NS) genes of a YF 17D infectious clone. The virus grew to high titers in cell cultures and was not neurovirulent for 3- to 4-week-old mice at doses </=6 log10 plaque forming units (pfu) inoculated by the intracerebral (IC) route. In contrast, commercial YF 17D vaccine was highly neurovirulent for weanling mice by the same route. Mice inoculated subcutaneously with one dose of >/=10(3) pfu of ChimeriVax-JE virus were solidly protected against intraperitoneal challenge with a virulent JE virus. Genetic stability of the chimera was assessed by sequential passages in cell cultures or in mouse brain. All attenuating residues and the avirulent phenotype were preserved after 18 passages in cell cultures or 6 passages in mouse brains.
Collapse
MESH Headings
- Animals
- Brain/virology
- Cell Line
- Chlorocebus aethiops
- Disease Models, Animal
- Encephalitis Virus, Japanese/genetics
- Encephalitis Virus, Japanese/immunology
- Encephalitis, Japanese/prevention & control
- Genes, Viral
- Genetic Vectors/genetics
- Genetic Vectors/physiology
- Humans
- Macaca mulatta
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred ICR
- Recombination, Genetic
- Sequence Analysis, DNA
- Vaccines, Attenuated/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vero Cells
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Virus Replication
- Yellow fever virus/genetics
- Yellow fever virus/growth & development
- Yellow fever virus/physiology
Collapse
Affiliation(s)
- F Guirakhoo
- OraVax, Inc., 38 Sidney Street, Cambridge, Massachusetts 02139, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Ryman KD, Ledger TN, Campbell GA, Watowich SJ, Barrett AD. Mutation in a 17D-204 vaccine substrain-specific envelope protein epitope alters the pathogenesis of yellow fever virus in mice. Virology 1998; 244:59-65. [PMID: 9581778 DOI: 10.1006/viro.1998.9057] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The heterogeneous nature of the yellow fever (YF) 17D-204 vaccine virus population was exploited in this study to isolate virus variants able to escape neutralization by the 17D-204 vaccine-specific MAb 864. The conformational change on the virus surface that resulted in the loss of the MAb 864-defined epitope was effected in each variant by a single amino acid mutation in the envelope (E) protein at either position E-305 or E-325. Interestingly, both positions were mutated during attenuation of the 17D-204 vaccine substrain from the wildtype Asibi strain. The mutations in several of the variants represented reversion to the wildtype Asibi virus sequence consistent with loss of a 17D-204 substrain-specific epitope. The majority of the variant viruses were shown to have altered mouse neurovirulence phenotypes, ranging from complete avirulence through to increased virulence. The avirulent variants are the first flavivirus MAb-neutralization-resistant variants to be attenuated for neurovirulence in the adult mouse model. Overall, the results indicate that the E protein epitope recognized by MAb 864 defines a functionally important region that encodes major molecular determinants of YF virus pathogenesis in vivo.
Collapse
Affiliation(s)
- K D Ryman
- Center for Tropical Diseases, Department of Pathology, University of Texas Medical Branch at Galveston, 77555, USA
| | | | | | | | | |
Collapse
|
37
|
Galler R, Freire MS, Jabor AV, Mann GF. The yellow fever 17D vaccine virus: molecular basis of viral attenuation and its use as an expression vector. Braz J Med Biol Res 1997; 30:157-68. [PMID: 9239300 DOI: 10.1590/s0100-879x1997000200002] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The yellow fever (YF) virus is the prototype flavivirus. The use of molecular techniques has unraveled the basic mechanisms of viral genome structure and expression. Recent trends in flavivirus research include the use of infectious clone technology with which it is possible to recover virus from cloned cDNA. Using this technique, mutations can be introduced at any point of the viral genome and their resulting effect on virus phenotype can be assessed. This approach has opened new possibilities to study several biological viral features with special emphasis on the issue of virulence/attenuation of the YF virus. The feasibility of using YF virus 17D vaccine strain, for which infectious cDNA is available, as a vector for the expression of heterologous antigens is reviewed.
Collapse
Affiliation(s)
- R Galler
- Laboratório de Tecnologia Virológica, Fundaçäo Instituto Oswaldo Cruz, Rio de Janeiro, Brasil.
| | | | | | | |
Collapse
|
38
|
Montefiori DC, Reimann KA, Letvin NL, Zhou J, Hu SL. Studies of complement-activating antibodies in the SIV/macaque model of acute primary infection and vaccine protection. AIDS Res Hum Retroviruses 1995; 11:963-70. [PMID: 7492443 DOI: 10.1089/aid.1995.11.963] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Questions regarding the potential impact of complement-activating antibodies on lentivirus pathogenesis and vaccine development were addressed in the SIV/macaque model by evaluating sera for activity related to complement-mediated, antibody-dependent enhancement (C'-ADE) of SIV infection in vitro. C'-ADE activity in sera obtained during acute primary infection in macaques inoculated with SIVmac251 appeared before neutralizing antibodies and coincided with the initial peak and decline of plasma antigenemia. The power of C'-ADE activity (i.e., virus production measured by p24 immunoassay) decreased as titers of neutralizing antibodies increased in these animals, suggesting a balance in the net effect between C'-ADE and neutralizing activities in vitro. Antibodies with C'-ADE activity were also induced in macaques immunized with live-attenuated SIVmac239/nef-deletion or primed with recombinant SIVmne gp120 vaccinia virus and boosted with SIVmne rgp160. The titer (i.e., last serum dilution to show enhancement), peak (i.e., serum dilution producing the greatest enhancement as measured by p24 production), and power (i.e., magnitude of p24 production at the peak titer) of C'-ADE activity in sera obtained from vaccinated macaques on the day of challenge were comparable to those of sera from infected macaques and showed no correlation with vaccine outcome, where some protected animals had C'-ADE profiles that resembled those of unprotected animals. The results of these studies suggest that antibodies having C'-ADE activity in vitro could contribute to virus replication or, alternatively, to virus clearance during the acute stage of SIV infection in macaques.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- D C Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
39
|
Gritsun TS, Holmes EC, Gould EA. Analysis of flavivirus envelope proteins reveals variable domains that reflect their antigenicity and may determine their pathogenesis. Virus Res 1995; 35:307-21. [PMID: 7785318 DOI: 10.1016/0168-1702(94)00090-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Studies on the molecular basis of flavivirus neutralisation, attenuation and tropism indicate that amino acid substitutions, in different parts of the envelope gene, may be responsible for the altered phenotypes. However, the association of particular substitutions with individual characteristics has proven difficult. Comparative analysis of all known tick-borne flavivirus envelope proteins through sequence alignment and a sliding window, reveals clusters of amino acid variation distributed throughout the envelope protein coding region. Further comparison with mosquito-borne flaviviruses reveals essentially the same profile of variability throughout the envelope protein sequence although there is a major difference within the postulated B domain of these viruses which may reflect their different evolutionary development. Most phenotypically variant properties, such as serotypic differences, variants characteristic of vaccine strains, altered tropisms and neutralisation escape mutants, map within the variable clusters. Thus, we propose that natural mutagenesis and selection may occur at specific sites that do not destroy the secondary and tertiary E protein structure and that the variable clusters represent the exposed surface amino acids of the envelope protein defining antigenicity, tropicity and pathogenesis.
Collapse
Affiliation(s)
- T S Gritsun
- NERC Institute of Virology and Environmental Microbiology, Oxford, UK
| | | | | |
Collapse
|
40
|
dos Santos CN, Post PR, Carvalho R, Ferreira II, Rice CM, Galler R. Complete nucleotide sequence of yellow fever virus vaccine strains 17DD and 17D-213. Virus Res 1995; 35:35-41. [PMID: 7754673 DOI: 10.1016/0168-1702(94)00076-o] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The complete nucleotide sequence of the genome from two yellow fever (YF) virus vaccine strains, 17DD and 17D-213, has been determined. Comparison of these sequences with those of other YF viruses including the parental virulent Asibi strain allowed the identification of 48 nucleotide sequence differences which are common to all 17D substrains. This is a significant reduction from the 67 nucleotide changes originally reported as being 17D-specific and potentially related to viral attenuation. The 48 changes are scattered throughout the genome, 26 of which are silent and 22 led to amino acid substitutions. These 22 changes are bona fide candidates to test by mutating the infectious YF cDNA to investigate their role in viral attenuation.
Collapse
Affiliation(s)
- C N dos Santos
- Fundação Oswaldo Cruz, Departamento de Bioquimica e Biologia Molecular, Rio de Janeiro, Brazil
| | | | | | | | | | | |
Collapse
|
41
|
Kawano H, Rostapshov V, Rosen L, Lai CJ. Genetic determinants of dengue type 4 virus neurovirulence for mice. J Virol 1993; 67:6567-75. [PMID: 8411360 PMCID: PMC238094 DOI: 10.1128/jvi.67.11.6567-6575.1993] [Citation(s) in RCA: 89] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Mouse-adapted dengue type 4 virus (DEN4) strain H241 is highly neurovirulent for mice, whereas its non-mouse-adapted parent is rarely neurovirulent. The genetic basis for the neurovirulence of the mouse-adapted mutant was studied by comparing intratypic chimeric viruses that contained the three structural protein genes from the parental virus or the neurovirulent mutant in the background sequence of nonneurovirulent DEN4 strain 814669. The chimera that contained the three structural protein genes from mouse neurovirulent DEN4 strain H241 proved to be highly neurovirulent in mice, whereas the chimera that contained the corresponding genes from its non-mouse-adapted parent was not neurovirulent. This finding indicates that most of the genetic loci for the neurovirulence of the DEN4 mutant lie within the structural protein genes. A comparison of the amino acid sequences of the parent and its mouse neurovirulent mutant proteins revealed that there were only five amino acid differences in the structural protein region, and three of these were located in the envelope (E) glycoprotein. Analysis of chimeras which contained one or two of the variant amino acids of the mutant E sequence substituting for the corresponding sequence of the parental virus identified two of these amino acid changes as important determinants of mouse neurovirulence. First, the single substitution of Ile for Thr-155 which ablated one of the two conserved glycosylation sites in parental E yielded a virus that was almost as neurovirulent as the mouse-adapted mutant. Thus, the loss of an E glycosylation site appears to play a role in DEN4 neurovirulence. Second, the substitution of Leu for Phe-401 also yielded a neurovirulent virus, but it was less neurovirulent than the glycosylation mutant. These findings indicate that at least two of the genetic loci responsible for DEN4 mouse neurovirulence map within the structural protein genes.
Collapse
Affiliation(s)
- H Kawano
- Molecular Viral Biology Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland 20892
| | | | | | | |
Collapse
|
42
|
Jennings AD, Whitby JE, Minor PD, Barrett AD. Comparison of the nucleotide and deduced amino acid sequences of the structural protein genes of the yellow fever 17DD vaccine strain from Senegal with those of other yellow fever vaccine viruses. Vaccine 1993; 11:679-81. [PMID: 8322493 DOI: 10.1016/0264-410x(93)90317-q] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The nucleic acid and deduced amino acid sequences of the structural proteins of the 17DD vaccine strain of yellow fever (YF) virus originating from Senegal are compared with those published for other vaccine strains of YF virus. Even though the 17D-204 and 17DD substrains of 17D diverged at passage 195 of 17D, they show a very high degree of nucleotide (99.5%) and amino acid (99.5%) homology over this region. The sequences are discussed with respect to monoclonal antibodies that can distinguish between 17DD and 17D-204 substrains of YF vaccines.
Collapse
Affiliation(s)
- A D Jennings
- Molecular Microbiology Group, School of Biological Sciences, University of Surrey, Guildford, UK
| | | | | | | |
Collapse
|