1
|
Zhao Z, Ke J, Liu M, Chen Z, Li S, Wei W, Mei K, Huang S. Pathogenicity and transmissibility differences in goose-originated subgenotype 3 duck Tembusu virus in goose embryos and 3-day-old goslings. Avian Pathol 2025; 54:175-184. [PMID: 39704145 DOI: 10.1080/03079457.2024.2373365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/23/2024] [Accepted: 06/22/2024] [Indexed: 12/21/2024]
Abstract
The Duck Tembusu virus (DTMUV) was first reported in China in 2010 and has since caused substantial economic losses in the poultry breeding industry. In the autumn of 2022, an outbreak of an infectious disease resembling DTMUV was reported in Guangdong Province, China, which caused significantly high mortality in goose embryos, and decreased egg production. This study identified one strain of the new subgenotype 3 of DTMUV, designated as DTMUV GDZQ2022, responsible for these effects. Comprehensive genomic sequencing of this strain was conducted to analyse its genetic variations. Additionally, the isolated and purified virus was inoculated into goose embryos and goslings to assess its pathogenicity. The GDZQ2022 genome displayed over 88% nucleotide homology with other DTMUV strains from China and Southeast Asia. Phylogenetic analysis of the E gene classified GDZQ2022 within the subgenotype 3 of DTMUV. Pathogenicity experiments on goose embryos and goslings showed that the GDZQ2022 strain induced typical clinical signs of DTMUV, particularly severe neurological manifestations. Although GDZQ2022 exhibited high virulence in goose embryos, its virulence in goslings was minimal, resulting in a low mortality rate. Pathological examinations detected significant histological lesions in the brains, livers, and spleens of the infected goslings. In conclusion, this study presents the first evidence of a novel DTMUV strain proliferating among young geese in China, underscoring the genetic diversity of DTMUV and contributing to our understanding of the pathogenicity of the subgenotype 3 Tembusu virus in goose embryos and goslings.RESEARCH HIGHLIGHTSPathogenicity of subgenotype 3 duck Tembusu virus in goose embryos and goslings was shown for the first time.DTMUV GDZQ2022 strain is highly pathogenic in goose embryos.The virulence of the DTMUV GDZQ2022 strain in goslings is relatively mild.Infected goslings exhibit typical clinical manifestations, with a low mortality rate.
Collapse
Affiliation(s)
- Ziqiao Zhao
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Junhong Ke
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Mengfan Liu
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Zuoxin Chen
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Shuwen Li
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Wuque Wei
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Kun Mei
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| | - Shujian Huang
- Waterfowl Infectious Disease Diagnosis and Prevention Research Team, School of Animal Science and Technology, Foshan University, Foshan, People's Republic of China
| |
Collapse
|
2
|
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
|
3
|
Identifying New Clusterons: Application of TBEV Analyzer 3.0. Microorganisms 2023; 11:microorganisms11020324. [PMID: 36838289 PMCID: PMC9966418 DOI: 10.3390/microorganisms11020324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/09/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Early knowledge about novel emerging viruses and rapid determination of their characteristics are crucial for public health. In this context, development of theoretical approaches to model viral evolution are important. The clusteron approach is a recent bioinformatics tool which analyzes genetic patterns of a specific E protein fragment and provides a hierarchical network structure of the viral population at three levels: subtype, lineage, and clusteron. A clusteron is a group of strains with identical amino acid (E protein fragment) signatures; members are phylogenetically closely related and feature a particular territorial distribution. This paper announces TBEV Analyzer 3.0, an analytical platform for rapidly characterizing tick-borne encephalitis virus (TBEV) strains based on the clusteron approach, workflow optimizations, and simplified parameter settings. Compared with earlier versions of TBEV Analyzer, we provide theoretical and practical enhancements to the platform. Regarding the theoretical aspect, the model of the clusteron structure, which is the core of platform analysis, has been updated by analyzing all suitable TBEV strains available in GenBank, while the practical enhancements aim at improving the platform's functionality. Here, in addition to expanding the strain sets of prior clusterons, we introduce eleven novel clusterons through our experimental results, predominantly of the European subtype. The obtained results suggest effective application of the proposed platform as an analytical and exploratory tool in TBEV surveillance.
Collapse
|
4
|
Erb SM, Butrapet S, Roehrig JT, Huang CYH, Blair CD. Genetic Adaptation by Dengue Virus Serotype 2 to Enhance Infection of Aedes aegypti Mosquito Midguts. Viruses 2022; 14:v14071569. [PMID: 35891549 PMCID: PMC9325310 DOI: 10.3390/v14071569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Dengue viruses (DENVs), serotypes 1–4, are arthropod-borne viruses transmitted to humans by mosquitoes, primarily Aedes aegypti. The transmission cycle begins when Ae. aegypti ingest blood from a viremic human and the virus infects midgut epithelial cells. In studying viruses derived from the DENV2 infectious clone 30P-NBX, we found that when the virus was delivered to female Ae. aegypti in an infectious blood meal, the midgut infection rate (MIR) was very low. To determine if adaptive mutations in the DENV2 envelope (E) glycoprotein could be induced to increase the MIR, we serially passed 30P-NBX in Ae. aegypti midguts. After four passages, a single, non-conservative mutation in E protein domain II (DII) nucleotide position 1300 became dominant, resulting in replacement of positively-charged amino acid lysine (K) at position 122 with negatively-charged glutamic acid (E; K122E) and a significantly-enhanced MIR. Site directed mutagenesis experiments showed that reducing the positive charge of this surface-exposed region of the E protein DII correlated with improved Ae. aegypti midgut infection.
Collapse
Affiliation(s)
- Steven M. Erb
- Center for Vector-Borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
| | - Siritorn Butrapet
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA; (S.B.); (J.T.R.); (C.Y.-H.H.)
| | - John T. Roehrig
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA; (S.B.); (J.T.R.); (C.Y.-H.H.)
| | - Claire Y.-H. Huang
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA; (S.B.); (J.T.R.); (C.Y.-H.H.)
| | - Carol D. Blair
- Center for Vector-Borne Infectious Diseases, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA;
- Correspondence:
| |
Collapse
|
5
|
Ku MW, Anna F, Souque P, Petres S, Prot M, Simon-Loriere E, Charneau P, Bourgine M. A Single Dose of NILV-Based Vaccine Provides Rapid and Durable Protection against Zika Virus. Mol Ther 2020; 28:1772-1782. [PMID: 32485138 PMCID: PMC7403329 DOI: 10.1016/j.ymthe.2020.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/30/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022] Open
Abstract
Zika virus, a member of the Flaviviridae family, is primarily transmitted by infected Aedes species mosquitoes. In 2016, Zika infection emerged as a global health emergency for its explosive spread and the remarkable neurological defects in the developing fetus. Development of a safe and effective Zika vaccine remains a high priority owing to the risk of re-emergence and limited understanding of Zika virus epidemiology. We engineered a non-integrating lentiviralvector(NILV)-based Zika vaccine encoding the consensus pre-membrane and envelope glycoprotein of circulating Zika virus strains. We further evaluated the immunogenicity and protective efficacy of this vaccine in both immunocompromised and immunocompetent mouse models. A single immunization in both mouse models elicited a robust neutralizing antibody titer and afforded full protection against Zika challenge as early as 7 days post-immunization. This NILV-based vaccine also induced a long-lasting immunity when immunized mice were challenged 6 months after immunization. Altogether, our NILV Zika vaccine provides a rapid yet durable protection through a single dose of immunization without extra adjuvant formulation. Our data suggest a promising Zika vaccine candidate for an emergency situation, and demonstrate the capacity of lentiviral vector as an efficient vaccine delivery platform.
Collapse
Affiliation(s)
- Min Wen Ku
- Unité de Virologie Moléculaire et Vaccinologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, 75005 Paris, France; Ecole Doctorale Frontières du Vivant (FdV), 26 Rue de l'Étoile, 75017 Paris, France
| | - François Anna
- Unité de Virologie Moléculaire et Vaccinologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Philippe Souque
- Unité de Virologie Moléculaire et Vaccinologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Stéphane Petres
- Plateforme Technologique Production et Purification de Protéines Recombinantes, Centre de Ressources et Recherches Technologiques, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Matthieu Prot
- Génomique Évolutive des Virus à ARN, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Etienne Simon-Loriere
- Génomique Évolutive des Virus à ARN, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Pierre Charneau
- Unité de Virologie Moléculaire et Vaccinologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France; Laboratoire commun Institut Pasteur-Theravectys, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France.
| | - Maryline Bourgine
- Unité de Virologie Moléculaire et Vaccinologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France; Laboratoire commun Institut Pasteur-Theravectys, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France.
| |
Collapse
|
6
|
Boelke M, Bestehorn M, Marchwald B, Kubinski M, Liebig K, Glanz J, Schulz C, Dobler G, Monazahian M, Becker SC. First Isolation and Phylogenetic Analyses of Tick-Borne Encephalitis Virus in Lower Saxony, Germany. Viruses 2019; 11:E462. [PMID: 31117224 PMCID: PMC6563265 DOI: 10.3390/v11050462] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis (TBE) is the most important tick-borne arboviral disease in Europe. Presently, the main endemic regions in Germany are located in the southern half of the country. Although recently, sporadic human TBE cases were reported outside of these known endemic regions. The detection and characterization of invading TBE virus (TBEV) strains will considerably facilitate the surveillance and assessment of this important disease. In 2018, ticks were collected by flagging in several locations of the German federal state of Lower Saxony where TBEV-infections in humans (diagnosed clinical TBE disease or detection of TBEV antibodies) were reported previously. Ticks were pooled according to their developmental stage and tested for TBEV-RNA by RT-qPCR. Five of 730 (0.68%) pools from Ixodes spp. ticks collected in the areas of "Rauher Busch" and "Barsinghausen/Mooshuette" were found positive for TBEV-RNA. Phylogenetic analysis of the whole genomes and E gene sequences revealed a close relationship between the two TBEV isolates, which cluster with a TBEV strain from Poland isolated in 1971. This study provides first data on the phylogeny of TBEV in the German federal state of Lower Saxony, outside of the known TBE endemic areas of Germany. Our results support the hypothesis of an east-west invasion of TBEV strains in Western Europe.
Collapse
Affiliation(s)
- Mathias Boelke
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| | - Malena Bestehorn
- Parasitology Unit, University of Hohenheim, Emil-Wolff-Straße 34, 70599 Stuttgart, Germany.
- Institute of Microbiology of the Bundeswehr, Neuherbergstraße 11, 80937 Munich, Germany.
| | - Birgit Marchwald
- The Governmental Institute of Public Health of Lower Saxony (NLGA), Roesebeckstraße 4-6, 30449 Hannover, Germany.
| | - Mareike Kubinski
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| | - Katrin Liebig
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| | - Julien Glanz
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| | - Claudia Schulz
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| | - Gerhard Dobler
- Parasitology Unit, University of Hohenheim, Emil-Wolff-Straße 34, 70599 Stuttgart, Germany.
- Institute of Microbiology of the Bundeswehr, Neuherbergstraße 11, 80937 Munich, Germany.
| | - Masyar Monazahian
- The Governmental Institute of Public Health of Lower Saxony (NLGA), Roesebeckstraße 4-6, 30449 Hannover, Germany.
| | - Stefanie C Becker
- Institute for Parasitology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
- Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hanover, Germany.
| |
Collapse
|
7
|
Klitting R, Roth L, Rey FA, de Lamballerie X. Molecular determinants of Yellow Fever Virus pathogenicity in Syrian Golden Hamsters: one mutation away from virulence. Emerg Microbes Infect 2018; 7:51. [PMID: 29593212 PMCID: PMC5874243 DOI: 10.1038/s41426-018-0053-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 02/07/2018] [Accepted: 02/11/2018] [Indexed: 11/12/2022]
Abstract
Yellow fever virus (Flavivirus genus) is an arthropod-borne pathogen, which can infect humans, causing a severe viscerotropic disease with a high mortality rate. Adapted viral strains allow the reproduction of yellow fever disease in hamsters with features similar to the human disease. Here, we used the Infectious Subgenomic Amplicons reverse genetics method to produce an equivalent to the hamster-virulent strain, Yellow Fever Ap7, by introducing a set of four synonymous and six nonsynonymous mutations into a single subgenomic amplicon, derived from the sequence of the Asibi strain. The resulting strain, Yellow Fever Ap7M, induced a disease similar to that described for Ap7 in terms of symptoms, weight evolution, viral loads in the liver and lethality. Using the same methodology, we produced mutant strains derived from either Ap7M or Asibi viruses and investigated the role of each of Ap7M nonsynonymous mutations in its in vivo phenotype. This allowed identifying key components of the virulence mechanism in hamsters. In Ap7M virus, the reversion of either E/Q27H or E/D155A mutations led to an important reduction of both virulence and in vivo replicative fitness. In addition, the introduction of the single D155A Ap7M mutation within the E protein of the Asibi virus was sufficient to drastically modify its phenotype in hamsters toward both a greater replication efficiency and virulence. Finally, inspection of the Asibi strain E protein structure combined to in vivo testing revealed the importance of an exposed α-helix in domain I, containing residues 154 and 155, for Ap7M virulence in hamsters.
Collapse
Affiliation(s)
- Raphaëlle Klitting
- UMR EPV: "Émergence des Pathologies Virales", Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP - IHU Méditerranée Infection, 13385, Marseille Cedex 05, France.
| | - Laura Roth
- UMR EPV: "Émergence des Pathologies Virales", Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP - IHU Méditerranée Infection, 13385, Marseille Cedex 05, France
| | - Félix A Rey
- Structural Virology Unit, Virology Department, Institut Pasteur, 75015, Paris, France
- CNRS UMR3569, Institut Pasteur, 75015, Paris, France
| | - Xavier de Lamballerie
- UMR EPV: "Émergence des Pathologies Virales", Aix-Marseille University - IRD 190 - Inserm 1207 - EHESP - IHU Méditerranée Infection, 13385, Marseille Cedex 05, France
| |
Collapse
|
8
|
Sun M, Dong J, Li L, Lin Q, Sun J, Liu Z, Shen H, Zhang J, Ren T, Zhang C. Recombinant Newcastle disease virus (NDV) expressing Duck Tembusu virus (DTMUV) pre-membrane and envelope proteins protects ducks against DTMUV and NDV challenge. Vet Microbiol 2018; 218:60-69. [PMID: 29685222 PMCID: PMC7117350 DOI: 10.1016/j.vetmic.2018.03.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/17/2022]
Abstract
Firstly generated a NDV-vectored Duck Tembusu Virus (DTMUV) bivalent vaccine that expressing the pre-membrane and envelope proteins of DTMUV. Evaluated the efficacy of the NDV-vectored Duck Tembusu Virus bivalent vaccine. Provided a new method for NDV and DTMUV controlling in waterfowl.
The newly emerged Duck Tembusu virus (DTMUV) is responsible for considerable economic loss in waterfowl-raising areas in China since 2010. Meanwhile, the virulent Newcastle disease virus (NDV) has also caused sporadic outbreaks in waterfowl. The individual vaccines against both diseases are available, however, there is no bivalent or combined vaccine for either disease. Here, we constructed a recombinant NDV-vectored vaccine candidate that expresses the pre-membrane (prM) and envelope (E) genes from DTMUV, designated as aGM/prM + E. The foreign prM and E proteins were stably expressed in aGM/prM + E and exhibited similar pathogenicity but higher growth kinetics than those of the parental virus. The aGM/prM + E carries a fusion cleavage site in accordance with avirulent viruses that have been frequently isolated from waterfowl, and induced remarkably (p < 0.001) higher NDV-specific hemagglutination inhibition (HI) titers than commercially available live NDV vaccines (LaSota strain). The aGM/prM + E also elicited significantly higher (p < 0.05) virus neutralization (VN) titers than commercially available DTMUV inactivated vaccines (HB strain). The aGM/prM + E not only provided complete protection against NDV challenge but also reduced the gross lesions on ovarian folliculi and provided 80% protection against DTMUV in ducks. We note that the aGM/prM + E vaccine can prevent challenged ducks from shedding of NDV and DTMUV. Our results suggest that the candidate vaccine aGM/prM + E would help decrease NDV and DTMUV transmissions in waterfowl raising areas in China.
Collapse
Affiliation(s)
- Minhua Sun
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Jiawen Dong
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Linlin Li
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Qiuyan Lin
- College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Guangzhou, Guangdong, China
| | - Junying Sun
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Zhicheng Liu
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Haiyan Shen
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Jianfeng Zhang
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China
| | - Tao Ren
- College of Veterinary Medicine, South China Agricultural University, 483 Wushan Road, Guangzhou, Guangdong, China.
| | - Chunhong Zhang
- Guangdong Provincial Key Laboratory of Livestock Disease Prevention; Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Techniques of Guangdong Province, Ministry of Agriculture; Guangdong Open Laboratory of Veterinary Public Health, Institute of Animal Health, Guangdong Academy of Agricultural Sciences, Baishigang Road, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Calvert AE, Dixon KL, Piper J, Bennett SL, Thibodeaux BA, Barrett ADT, Roehrig JT, Blair CD. A humanized monoclonal antibody neutralizes yellow fever virus strain 17D-204 in vitro but does not protect a mouse model from disease. Antiviral Res 2016; 131:92-9. [PMID: 27126613 PMCID: PMC4899248 DOI: 10.1016/j.antiviral.2016.04.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 04/20/2016] [Accepted: 04/23/2016] [Indexed: 01/29/2023]
Abstract
The yellow fever virus (YFV) vaccine 17D-204 is considered safe and effective, yet rare severe adverse events (SAEs), some resulting in death, have been documented following vaccination. Individuals exhibiting post-vaccinal SAEs are ideal candidates for antiviral monoclonal antibody (MAb) therapy; the time until appearance of clinical signs post-exposure is usually short and patients are quickly hospitalized. We previously developed a murine-human chimeric monoclonal antibody (cMAb), 2C9-cIgG, reactive with both virulent YFV and 17D-204, and demonstrated its ability to prevent and treat YF disease in both AG129 mouse and hamster models of infection. To counteract possible selection of 17D-204 variants that escape neutralization by treatment with a single MAb (2C9-cIgG), we developed a second cMAb, 864-cIgG, for use in combination with 2C9-cIgG in post-vaccinal therapy. MAb 864-cIgG recognizes/neutralizes only YFV 17D-204 vaccine substrain and binds to domain III (DIII) of the viral envelope protein, which is different from the YFV type-specific binding site of 2C9-cIgG in DII. Although it neutralized 17D-204 in vitro, administration of 864-cIgG had no protective capacity in the interferon receptor-deficient AG129 mouse model of 17D-204 infection. The data presented here show that although DIII-specific 864-cIgG neutralizes virus infectivity in vitro, it does not have the ability to abrogate disease in vivo. Therefore, combination of 864-cIgG with 2C9-cIgG for treatment of YF vaccination SAEs does not appear to provide an improvement on 2C9-cIgG therapy alone.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Disease Models, Animal
- Humans
- Immunization, Passive
- Mice
- Neutralization Tests
- Receptors, Interferon/deficiency
- Receptors, Interferon/genetics
- Viral Envelope Proteins/immunology
- Viral Envelope Proteins/metabolism
- Yellow Fever/immunology
- Yellow Fever/prevention & control
- Yellow Fever/therapy
- Yellow Fever Vaccine/adverse effects
- Yellow Fever Vaccine/immunology
- Yellow fever virus/immunology
Collapse
Affiliation(s)
- Amanda E Calvert
- Division of Vector-Borne Diseases, U.S. Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Kandice L Dixon
- Division of Vector-Borne Diseases, U.S. Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Joseph Piper
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, 80523-1692, USA
| | - Susan L Bennett
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, 80523-1692, USA
| | - Brett A Thibodeaux
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, 80523-1692, USA
| | - Alan D T Barrett
- Department of Pathology and Sealy Center for Vaccine Development, University of Texas-Medical Branch, Galveston, TX, 77555, USA
| | - John T Roehrig
- Division of Vector-Borne Diseases, U.S. Centers for Disease Control and Prevention, Fort Collins, CO, 80521, USA
| | - Carol D Blair
- Arthropod-borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, 80523-1692, USA.
| |
Collapse
|
10
|
Nikolay B, Fall G, Boye CSB, Sall AA, Skern T. Validation of a structural comparison of the antigenic characteristics of Usutu virus and West Nile virus envelope proteins. Virus Res 2014; 189:87-91. [PMID: 24874193 DOI: 10.1016/j.virusres.2014.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 10/25/2022]
Abstract
Cross-reactions observed in serological assays between Usutu virus (USUV), the USUV outlier subtype strain CAR_1969 and West Nile virus (WNV) suggest that they share antigenic features amongst their structural outer proteins especially envelope (E) proteins. To investigate the molecular background of this observation, we compared the E protein sequences of seven USUV strains, USUV subtype strain CAR_1969 and WNV strain 2471, focusing on the binding site defined by the WNV neutralizing antibody E16. USUV SouthAfrica_1959 differs from WNV 2741 in three of four residues critical for E16 antibody binding and five of the 12 additionally involved residues. In contrast, USUV subtype CAR_1969 differs from WNV 2741 in two critical residues and five additional residues. Furthermore, USUV subtype CAR_1969 differs from other USUV strains in two critical residues. E16 antibody binding has previously been shown to be highly specific for WNV; thus, the observed variation in amino acid residues suggests that the region corresponding to the WNV E16 epitope is probably not responsible for the observed cross-reactions between WNV and USUV. Seroneutralisation assays confirmed these findings for WNV and USUV, however, showed occurring cross-reactivity between WNV and USUV subtype CAR_1969 at high antibody titers. The sequence diversity in this region might also explain some of the observed different antigenic characteristics of USUV strains and USUV subtype CAR_1969. A therapeutic effect of E16 antibody has been described in WNV infected mice; therefore, a USUV specific antibody generated against the region corresponding to the WNV E16 binding site might represent an approach for treating USUV infections.
Collapse
Affiliation(s)
- Birgit Nikolay
- Unité des arbovirus et virus de fièvres hémorragiques, Institut Pasteur de Dakar, 36 Avenue Pasteur, BP 220 Dakar, Senegal; Faculty of Life Sciences, University of Vienna, Dr. Bohr-Gasse 9/3, A-1030 Vienna, Austria; Faculté de Médecine, Pharmacie, Odonto-stomatologie, Université Cheikh Anta Diop Dakar, 24 Avenue Cheikh Anta Diop, Dakar, Senegal.
| | - Gamou Fall
- Unité des arbovirus et virus de fièvres hémorragiques, Institut Pasteur de Dakar, 36 Avenue Pasteur, BP 220 Dakar, Senegal.
| | - Cheikh Saad Bouh Boye
- Faculté de Médecine, Pharmacie, Odonto-stomatologie, Université Cheikh Anta Diop Dakar, 24 Avenue Cheikh Anta Diop, Dakar, Senegal.
| | - Amadou Alpha Sall
- Unité des arbovirus et virus de fièvres hémorragiques, Institut Pasteur de Dakar, 36 Avenue Pasteur, BP 220 Dakar, Senegal.
| | - Tim Skern
- Max F. Perutz Laboratories, Medical University of Vienna, Dr. Bohr-Gasse 9/3, A-1030 Vienna, Austria.
| |
Collapse
|
11
|
Suzarte E, Marcos E, Gil L, Valdés I, Lazo L, Ramos Y, Pérez Y, Falcón V, Romero Y, Guzmán MG, González S, Kourí J, Guillén G, Hermida L. Generation and characterization of potential dengue vaccine candidates based on domain III of the envelope protein and the capsid protein of the four serotypes of dengue virus. Arch Virol 2014; 159:1629-40. [DOI: 10.1007/s00705-013-1956-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023]
|
12
|
Vratskikh O, Stiasny K, Zlatkovic J, Tsouchnikas G, Jarmer J, Karrer U, Roggendorf M, Roggendorf H, Allwinn R, Heinz FX. Dissection of antibody specificities induced by yellow fever vaccination. PLoS Pathog 2013; 9:e1003458. [PMID: 23818856 PMCID: PMC3688551 DOI: 10.1371/journal.ppat.1003458] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 05/11/2013] [Indexed: 12/30/2022] Open
Abstract
The live attenuated yellow fever (YF) vaccine has an excellent record of efficacy and one dose provides long-lasting immunity, which in many cases may last a lifetime. Vaccination stimulates strong innate and adaptive immune responses, and neutralizing antibodies are considered to be the major effectors that correlate with protection from disease. Similar to other flaviviruses, such antibodies are primarily induced by the viral envelope protein E, which consists of three distinct domains (DI, II, and III) and is presented at the surface of mature flavivirions in an icosahedral arrangement. In general, the dominance and individual variation of antibodies to different domains of viral surface proteins and their impact on neutralizing activity are aspects of humoral immunity that are not well understood. To gain insight into these phenomena, we established a platform of immunoassays using recombinant proteins and protein domains that allowed us to dissect and quantify fine specificities of the polyclonal antibody response after YF vaccination in a panel of 51 vaccinees as well as determine their contribution to virus neutralization by serum depletion analyses. Our data revealed a high degree of individual variation in antibody specificities present in post-vaccination sera and differences in the contribution of different antibody subsets to virus neutralization. Irrespective of individual variation, a substantial proportion of neutralizing activity appeared to be due to antibodies directed to complex quaternary epitopes displayed on the virion surface only but not on monomeric E. On the other hand, DIII-specific antibodies (presumed to have the highest neutralizing activity) as well as broadly flavivirus cross-reactive antibodies were absent or present at very low titers. These data provide new information on the fine specificity as well as variability of antibody responses after YF vaccination that are consistent with a strong influence of individual-specific factors on immunodominance in humoral immune responses. The live-attenuated yellow fever vaccine has been administered to more than 600 million people worldwide and is considered to be one of the most successful viral vaccines ever produced. Following injection, the apathogenic vaccine virus replicates in the vaccinee and induces antibodies that mediate virus neutralization and subsequent protection from disease. In principle, many different antibodies are induced by viral antigens, but it is becoming increasingly clear that only a subset of them is capable of inactivating the virus, and some antibody populations appear to dominate the immune response. However, to date there has been very little information on individual-specific variations of immunodominance and how such variations can affect the functionality of antibody responses. In our study, we addressed these issues and analyzed the fine specificities of antibodies induced by YF vaccination as well as the contribution of different antibody subsets to virus neutralization in 51 vaccinees. We demonstrate an extensive degree of individual variation with respect to immunodominance of antibody populations and their contribution to virus neutralization. Such variations can have an impact on vaccine-mediated protection, and thus insight into this phenomenon can provide leads for novel strategies in modern vaccine design.
Collapse
Affiliation(s)
- Oksana Vratskikh
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Jürgen Zlatkovic
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | | | - Johanna Jarmer
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Urs Karrer
- Division of Infectious Diseases, University Hospital of Zurich, Zurich, Switzerland
| | | | - Hedwig Roggendorf
- Institute for Virology, University of Duisburg-Essen, Essen, Germany
| | - Regina Allwinn
- Institute for Medical Virology, University Hospital Frankfurt am Main, Frankfurt am Main, Germany
| | - Franz X. Heinz
- Department of Virology, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
13
|
Kollaritsch H, Paulke-Korinek M, Holzmann H, Hombach J, Bjorvatn B, Barrett A. Vaccines and vaccination against tick-borne encephalitis. Expert Rev Vaccines 2013; 11:1103-19. [PMID: 23151167 DOI: 10.1586/erv.12.86] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Tick-borne encephalitis (TBE) is an emerging viral zoonosis and is endemic from Japan, China, Mongolia and Russia, to Central Europe and France. There is no specific treatment and TBE can be fatal. The four licensed prophylactic vaccines are produced according to WHO manufacturing requirements. Large clinical trials and postmarketing surveillance demonstrated safety and efficacy of the two European vaccines. The two Russian vaccines showed their effectiveness in daily use, but limited published data are available on controlled clinical trials. Vaccination recommendations in endemic areas vary significantly. In some countries, public vaccination programs are implemented. The WHO has recently issued recommendations on evidence-based use of TBE vaccines. However, more data are needed regarding safety, efficacy and long-term protection after vaccination.
Collapse
Affiliation(s)
- Herwig Kollaritsch
- Department of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Kinderspitalgasse 15, Vienna, Austria
| | | | | | | | | | | |
Collapse
|
14
|
|
15
|
Heinz F, Stiasny K. Flaviviruses and their antigenic structure. J Clin Virol 2012; 55:289-95. [DOI: 10.1016/j.jcv.2012.08.024] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 08/25/2012] [Indexed: 12/13/2022]
|
16
|
Testa JS, Shetty V, Sinnathamby G, Nickens Z, Hafner J, Kamal S, Zhang X, Jett M, Philip R. Conserved MHC class I-presented dengue virus epitopes identified by immunoproteomics analysis are targets for cross-serotype reactive T-cell response. J Infect Dis 2012; 205:647-55. [PMID: 22246683 DOI: 10.1093/infdis/jir814] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Dengue fever and dengue hemorrhagic fever are significant global public health problems, and understanding the overall immune response to infection will contribute to appropriate management of the disease and its potentially severe complications. Live attenuated and subunit vaccine candidates, which are under clinical evaluation, induce primarily an antibody response to the virus and minimal cross-reactive T-cell responses. Currently, there are no available tools to assess protective T-cell responses during infection or after vaccination. In this study, we utilize an immunoproteomics process to uncover novel HLA-A2-specific epitopes derived from dengue virus (DV)-infected cells. These epitopes are conserved, and we report that epitope-specific cytotoxic lymphocytes (CTLs) are cross-reactive against all 4 DV serotypes. These epitopes have potential as new informational and diagnostic tools to characterize T-cell immunity in DV infection and may serve as part of a universal vaccine candidate complementary to current vaccines in trial.
Collapse
Affiliation(s)
- James S Testa
- Immunotope Inc, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Enhancing the utility of a prM/E-expressing chimeric vaccine for Japanese encephalitis by addition of the JEV NS1 gene. Vaccine 2011; 29:7444-55. [PMID: 21798299 DOI: 10.1016/j.vaccine.2011.07.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 07/08/2011] [Accepted: 07/17/2011] [Indexed: 12/11/2022]
Abstract
Recently, we demonstrated that a single-cycle West Nile virus (WNV) named RepliVAX WN could be used to produce a chimeric Japanese encephalitis (JE) vaccine (RepliVAX JE) by replacing the WNV prM/E genes with those of JEV. Here, we tested if replacement of WNV NS1 gene in RepliVAX JE with that of JEV (producing TripliVAX JE) could produce a superior vaccine. TripliVAX JE elicited higher anti-E immunity and displayed better efficacy in mice than RepliVAX JE. Furthermore, TripliVAX JE displayed reduced immune interference caused by pre-existing anti-NS1 immunity. Thus, we propose prM/E/NS1 chimerization as a new strategy for flavivirus vaccine development.
Collapse
|
18
|
Gangwar RS, Shil P, Cherian SS, Gore MM. Delineation of an epitope on domain I of Japanese encephalitis virus Envelope glycoprotein using monoclonal antibodies. Virus Res 2011; 158:179-87. [PMID: 21477626 DOI: 10.1016/j.virusres.2011.03.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 03/29/2011] [Accepted: 03/30/2011] [Indexed: 12/22/2022]
Abstract
The Envelope glycoprotein (E-protein) of Japanese encephalitis virus (JEV) is the major structural component on the virion surface and is a primary target for the host immune system. Two monoclonal antibodies (MAbs) NHA-I (IgG2b) and NHA-II (IgM) against JEV (Indian strain 733913) were earlier developed in the authors' laboratory and found to be cross-reactive to nuclear histones. However, the epitope specificity of these MAbs has remained unknown. The present study was carried out to delineate the epitopes recognised by these MAbs on the E-protein of JEV strain 733913. The variable regions of the NHA-I and NHA-II were sequenced and the tertiary structures predicted. Molecular docking of the MAbs with the structural model of the JEV E-protein demonstrated that NHA-I binds to a predicted antigenic determinant (residue position 18-33) in domain-I. To understand the epitope specificity and check for possible cross-reactivity of these MAbs, comparative analysis of interactions with the known crystallographic structure of the West Nile virus (WNV) E-protein was also carried out. The studies predicted a differential binding of NHA-I but not of NHA-II between JEV and WNV. Mutagenesis studies could help analyse the specificity of NHA-I. The NHA-II appears to be cross-reactive as it docked in the groove region between domains I and III of both the JEV and WNV E-proteins. In laboratory assays, namely, ELISA and immunofluorescence assay both the MAbs reacted equally with JEV while the NHA-I did not show any reactivity with WNV. In silico results were thus validated by laboratory experiments. The present study would help in better understanding of virus-host interactions at the molecular level, and also be useful for the future design of vaccines as well as peptide based diagnostics.
Collapse
Affiliation(s)
- Roopesh Singh Gangwar
- Japanese Encephalitis Group, National Institute of Virology, Sus Road Campus, 130/1, Pashan, Pune 411021, India
| | | | | | | |
Collapse
|
19
|
Ding T, Zhang W, Ma W, Ren J. Identification of a mutated BHK-21 cell line that became less susceptible to Japanese encephalitis virus infection. Virol J 2011; 8:115. [PMID: 21396132 PMCID: PMC3064645 DOI: 10.1186/1743-422x-8-115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 03/14/2011] [Indexed: 04/23/2025] Open
Abstract
The pathogenesis of Japanese encephalitis virus (JEV) is not definitely elucidated as the initial interaction between virus and host cell receptors required for JEV infection is not clearly defined yet. Here, in order to discover those membrane proteins that may be involved in JEV attachment to or entry into virus permissive BHK-21 cells, a chemically mutated cell line (designated 3A10-3F) that became less susceptible to JEV infection was preliminarily established and selected by repeated low moi JEV challenges and RT-PCR detection for viral RNA E gene fragment. The susceptibility to JEV of 3A10-3F cells was significantly weakened compared with parental BHK-21 cells, verified by indirect immunofluorescence assay, virus plague formation assay, and flow cytometry. Finally, two-dimensional electrophoresis (2-DE) coupled with LC-MS/MS was utilized to recognize the most differentially expressed proteins from membrane protein extracts of 3A10-3F and BHK-21 cells respectively. The noted discrepancy of membrane proteins included calcium binding proteins (annexin A1, annexin A2), and voltage-dependent anion channels proteins (VDAC 1, VDAC 2), suggesting that these molecules may affect JEV attachment to and/or entry into BHK-21 cells and worthy of further investigation.
Collapse
Affiliation(s)
- Tianbing Ding
- Department of Microbiology, the Fourth Military Medical University, Xi'an, Shaanxi, 710032, PR China
| | | | | | | |
Collapse
|
20
|
Umamaheswari A, Kumar MM, Pradhan D, Marisetty H. Docking studies towards exploring antiviral compounds against envelope protein of yellow fever virus. Interdiscip Sci 2011; 3:64-77. [PMID: 21369890 DOI: 10.1007/s12539-011-0064-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 06/04/2010] [Accepted: 06/07/2010] [Indexed: 10/18/2022]
Abstract
Yellow fever is among one of the most lethal viral diseases for which approved antiviral therapies were yet to be discovered. Herein, functional assignment of complete YFV proteome was done through support vector machine. Major envelope (E) protein that mediates entry of YFV into host cell was selected as a potent molecular target. Three dimensional structure of the molecular target was predicted using Modeller9v7. The model was optimized in Maestro9.0 applying OPLS AA force field and was evaluated using PROCHECK, ProSA, ProQ and Profile 3D. The BOG pocket residues Val48, Glu197, Thr200, Ile204, Thr265, Thr268 and Gly278 were located in YFV E protein using SiteMap2.3. More than one million compounds of Ligandinfo Meta database were explored using a computational virtual screening protocol targeting BOG pocket of the E protein. Finally, ten top ranked lead molecules with strong binding affinity to BOG pocket of YFV E protein were identified based on XP Gscore. Drug likeliness and comparative bioactivity analysis for these leads using QikProp3.2 had shown that these molecules would have the potential to act as better drug. Thus, the 10 lead molecules suggested in the present study would be of interest as promising starting point for designing antiviral compound against yellow fever.
Collapse
Affiliation(s)
- Amineni Umamaheswari
- SVIMS Bioinformatics Centre, Department of Bioinformatics, SVIMS University, Tirupati, 517507, AP, India.
| | | | | | | |
Collapse
|
21
|
Butrapet S, Childers T, Moss KJ, Erb SM, Luy BE, Calvert AE, Blair CD, Roehrig JT, Huang CYH. Amino acid changes within the E protein hinge region that affect dengue virus type 2 infectivity and fusion. Virology 2011; 413:118-27. [PMID: 21353281 DOI: 10.1016/j.virol.2011.01.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 12/12/2010] [Accepted: 01/24/2011] [Indexed: 10/18/2022]
Abstract
Fifteen mutant dengue viruses were engineered and used to identify AAs in the molecular hinge of the envelope protein that are critical to viral infection. Substitutions at Q52, A54, or E133 reduced infectivity in mammalian cells and altered the pH threshold of fusion. Mutations at F193, G266, I270, or G281 affected viral replication in mammalian and mosquito cells, but only I270W had reduced fusion activity. T280Y affected the pH threshold for fusion and reduced replication in C6/36 cells. Three different mutations at L135 were lethal in mammalian cells. Among them, L135G abrogated fusion and reduced replication in C6/36 cells, but only slightly reduced the mosquito infection rate. Conversely, L135W replicated well in C6/36 cells, but had the lowest mosquito infection rate. Possible interactions between hinge residues 52 and 277, or among 53, 135, 170, 186, 265, and 276 required for hinge function were discovered by sequence analysis to identify compensatory mutations.
Collapse
Affiliation(s)
- Siritorn Butrapet
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, CO 80521, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Domain-III FG loop of the dengue virus type 2 envelope protein is important for infection of mammalian cells and Aedes aegypti mosquitoes. Virology 2010; 406:328-35. [DOI: 10.1016/j.virol.2010.07.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Revised: 06/22/2010] [Accepted: 07/18/2010] [Indexed: 11/24/2022]
|
23
|
A novel mutation (S227T) in domain II of the envelope gene of Japanese encephalitis virus circulating in North India. Epidemiol Infect 2010; 139:849-56. [PMID: 20727244 DOI: 10.1017/s0950268810001937] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Japanese encephalitis (JE) is an important arboviral infection of public health concern. There is a significant variation in mortality (10-30%) in JE viral infection. Epidemics of JE have become regular features in the northern states of India. The recent resurgence of the A226V mutation leading to a widespread Chikungunya epidemic motivated the investigators to search for any such mutational occurrence with Japanese encephalitis virus (JEV) isolated from this region. This study looked for mutation of clinical strains at amino-acid positions 176, 177, 227, 244, 264 and 279. A novel mutation S227T was detected corresponding to the loop region of domain II, E gene of JEV in comparison to Indian and other isolates from different parts of the world. Genotype III was found to be circulating in this geographical area. Further studies are required to ascertain its role in JE pathogenesis and vector competency.
Collapse
|
24
|
Kimura T, Sasaki M, Okumura M, Kim E, Sawa H. Flavivirus encephalitis: pathological aspects of mouse and other animal models. Vet Pathol 2010; 47:806-18. [PMID: 20551474 DOI: 10.1177/0300985810372507] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Encephalitic flaviviruses are important arthropod-borne pathogens of humans and other animals. In particular, the recent emergence of the West Nile virus (WNV) and Japanese encephalitis virus (JEV) in new geographic areas has caused a considerable public health alert and international concern. Among the experimental in vivo models of WNV and JEV infection, mice and other laboratory rodents are the most thoroughly studied and well-characterized systems, having provided data that are important for understanding the infectious process in humans. Macaca monkeys have also been used as a model for WNV and JEV infection, mainly for the evaluation of vaccine efficacy, although a limited number of published studies have addressed pathomorphology. These animal models demonstrate the development of encephalitis with many similarities to the human disease; however, the histological events that occur during infection, especially in peripheral tissues, have not been fully characterized.
Collapse
Affiliation(s)
- T Kimura
- Department of Molecular Pathobiology, Hokkaido University Research Center for Zoonosis Control, West 10 North 20, Kita-ku, Sapporo 001-0020 Japan.
| | | | | | | | | |
Collapse
|
25
|
JNK phosphorylation, induced during dengue virus infection, is important for viral infection and requires the presence of cholesterol. Virology 2010; 396:30-6. [DOI: 10.1016/j.virol.2009.10.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2009] [Revised: 07/27/2009] [Accepted: 10/13/2009] [Indexed: 01/07/2023]
|
26
|
Huang CYH, Butrapet S, Moss KJ, Childers T, Erb SM, Calvert AE, Silengo SJ, Kinney RM, Blair CD, Roehrig JT. The dengue virus type 2 envelope protein fusion peptide is essential for membrane fusion. Virology 2009; 396:305-15. [PMID: 19913272 DOI: 10.1016/j.virol.2009.10.027] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 09/28/2009] [Accepted: 10/16/2009] [Indexed: 10/20/2022]
Abstract
The flaviviral envelope (E) protein directs virus-mediated membrane fusion. To investigate membrane fusion as a requirement for virus growth, we introduced 27 unique mutations into the fusion peptide of an infectious cDNA clone of dengue 2 virus and recovered seven stable mutant viruses. The fusion efficiency of the mutants was impaired, demonstrating for the first time the requirement for specific FP AAs in optimal fusion. Mutant viruses exhibited different growth kinetics and/or genetic stabilities in different cell types and adult mosquitoes. Virus particles could be recovered following RNA transfection of cells with four lethal mutants; however, recovered viruses could not re-infect cells. These viruses could enter cells, but internalized virus appeared to be retained in endosomal compartments of infected cells, thus suggesting a fusion blockade. Mutations of the FP also resulted in reduced virus reactivity with flavivirus group-reactive antibodies, confirming earlier reports using virus-like particles.
Collapse
Affiliation(s)
- Claire Y-H Huang
- Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Public Health Service, U.S. Department of Health and Human Services, 3150 Rampart Rd., Fort Collins, CO 80521, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hall RA, Tan SE, Selisko B, Slade R, Hobson-Peters J, Canard B, Hughes M, Leung JY, Balmori-Melian E, Hall-Mendelin S, Pham KB, Clark DC, Prow NA, Khromykh AA. Monoclonal antibodies to the West Nile virus NS5 protein map to linear and conformational epitopes in the methyltransferase and polymerase domains. J Gen Virol 2009; 90:2912-2922. [PMID: 19710254 DOI: 10.1099/vir.0.013805-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The West Nile virus (WNV) NS5 protein contains a methyltransferase (MTase) domain involved in RNA capping and an RNA-dependent RNA polymerase (RdRp) domain essential for virus replication. Crystal structures of individual WNV MTase and RdRp domains have been solved; however, the structure of full-length NS5 has not been determined. To gain more insight into the structure of NS5 and interactions between the MTase and RdRp domains, we generated a panel of seven monoclonal antibodies (mAbs) to the NS5 protein of WNV (Kunjin strain) and mapped their binding sites using a series of truncated NS5 proteins and synthetic peptides. Binding sites of four mAbs (5D4, 4B6, 5C11 and 6A10) were mapped to residues 354-389 in the fingers subdomain of the RdRp. This is consistent with the ability of these mAbs to inhibit RdRp activity in vitro and suggests that this region represents a potential target for RdRp inhibitors. Using a series of synthetic peptides, we also identified a linear epitope (bound by mAb 5H1) that mapped to a 13 aa stretch surrounding residues 47 and 49 in the MTase domain, a region predicted to interact with the palm subdomain of the RdRp. The failure of one mAb (7G6) to bind both N- and C-terminally truncated NS5 recombinants indicates that the antibody recognizes a conformational epitope that requires the presence of residues in both the MTase and RdRp domains. These data support a structural model of the full-length NS5 molecule that predicts a physical interaction between the MTase and the RdRp domains.
Collapse
Affiliation(s)
- Roy A Hall
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Si En Tan
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Barbara Selisko
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique, Marseille, France
| | - Rachael Slade
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Jody Hobson-Peters
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Bruno Canard
- Architecture et Fonction des Macromolécules Biologiques, Centre National de la Recherche Scientifique, Marseille, France
| | - Megan Hughes
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Jason Y Leung
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Ezequiel Balmori-Melian
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Sonja Hall-Mendelin
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Kim B Pham
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - David C Clark
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Natalie A Prow
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Alexander A Khromykh
- Centre for Infectious Desease Research, School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
28
|
Ishikawa T, Widman DG, Bourne N, Konishi E, Mason PW. Construction and evaluation of a chimeric pseudoinfectious virus vaccine to prevent Japanese encephalitis. Vaccine 2008; 26:2772-81. [PMID: 18433947 DOI: 10.1016/j.vaccine.2008.03.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Revised: 03/01/2008] [Accepted: 03/11/2008] [Indexed: 11/17/2022]
Abstract
Multiple vaccines exist to control Japanese encephalitis (JE), but all suffer from problems. We have developed a new type of flavivirus vaccine, a pseudoinfectious virus (RepliVAX WN) that prevents West Nile virus (WNV)-induced disease. Here, we describe production of a chimeric RepliVAX (RepliVAX JE) that expresses the JE virus (JEV) prM and E proteins. Our prototype RepliVAX JE replicated poorly in cells, but blind passage produced a better-growing derivative, and analyses of this derivative allowed us to engineer a second-generation RepliVAX (RepliVAX JE.2) that grew to high titers. RepliVAX JE.2 elicited neutralizing antibodies in both mice and hamsters and provided 100% protection from a lethal challenge with JEV or WNV, respectively. These results demonstrate the utility our RepliVAX platform for producing a JE vaccine.
Collapse
Affiliation(s)
- Tomohiro Ishikawa
- Department of Pathology, University of Texas Medical Branch (UTMB), 301 University Boulevard, Galveston, TX 77555-0436, United States
| | | | | | | | | |
Collapse
|
29
|
Marasco WA, Sui J. The growth and potential of human antiviral monoclonal antibody therapeutics. Nat Biotechnol 2008; 25:1421-34. [PMID: 18066039 PMCID: PMC7097443 DOI: 10.1038/nbt1363] [Citation(s) in RCA: 210] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Monoclonal antibodies (mAbs) have long provided powerful research tools for virologists to understand the mechanisms of virus entry into host cells and of antiviral immunity. Even so, commercial development of human (or humanized) mAbs for the prophylaxis, preemptive and acute treatment of viral infections has been slow. This is surprising, as new antibody discovery tools have increased the speed and precision with which potent neutralizing human antiviral mAbs can be identified. As longstanding barriers to antiviral mAb development, such as antigenic variability of circulating viral strains and the ability of viruses to undergo neutralization escape, are being overcome, deeper insight into the mechanisms of mAb action and engineering of effector functions are also improving the efficacy of antiviral mAbs. These successes, in both industrial and academic laboratories, coupled with ongoing changes in the biomedical and regulatory environments, herald an era when the commercial development of human antiviral mAb therapies will likely surge.
Collapse
Affiliation(s)
- Wayne A Marasco
- Department of Cancer Immunology & AIDS, Dana-Farber Cancer Institute and Department of Medicine, Harvard Medical School 44, Binney Street, Boston, Massachusetts 02115, USA.
| | | |
Collapse
|
30
|
|
31
|
Lai CJ, Goncalvez AP, Men R, Wernly C, Donau O, Engle RE, Purcell RH. Epitope determinants of a chimpanzee dengue virus type 4 (DENV-4)-neutralizing antibody and protection against DENV-4 challenge in mice and rhesus monkeys by passively transferred humanized antibody. J Virol 2007; 81:12766-74. [PMID: 17881450 PMCID: PMC2169078 DOI: 10.1128/jvi.01420-07] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The chimpanzee monoclonal antibody (MAb) 5H2 is specific for dengue virus type 4 (DENV-4) and neutralizes the virus at a high titer in vitro. The epitope detected by the antibody was mapped by sequencing neutralization escape variants of the virus. One variant contained a Lys174-Glu substitution and another contained a Pro176-Leu substitution in domain I of the DENV-4 envelope protein (E). These mutations reduced binding affinity for the antibody 18- to >100-fold. Humanized immunoglobulin G (IgG) 5H2, originally produced from an expression vector, has been shown to be a variant containing a nine-amino-acid deletion in the Fc region which completely ablates antibody-dependent enhancement of DENV replication in vitro. The variant MAb, termed IgG 5H2 deltaD, is particularly attractive for exploring its protective capacity in vivo. Passive transfer of IgG 5H2 deltaD at 20 microg/mouse afforded 50% protection of suckling mice against challenge with 25 50% lethal doses of mouse neurovirulent DENV-4 strain H241. Passive transfer of antibody to monkeys was conducted to demonstrate proof of concept for protection against DENV challenge. Monkeys that received 2 mg/kg of body weight of IgG 5H2 deltaD were completely protected against 100 50% monkey infectious doses (MID50) of DENV-4, as indicated by the absence of viremia and seroconversion. A DENV-4 escape mutant that contained a Lys174-Glu substitution identical to that found in vitro was isolated from monkeys challenged with 10(6) MID50 of DENV-4. This substitution was also present in all naturally occurring isolates belonging to DENV-4 genotype III. These studies have important implications for possible antibody-mediated prevention of DENV infection.
Collapse
Affiliation(s)
- Ching-Juh Lai
- Laboratory of Infectious Diseases, National Institutes of Health, 50 South Drive MSC 8005, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Sukupolvi-Petty S, Austin SK, Purtha WE, Oliphant T, Nybakken GE, Schlesinger JJ, Roehrig JT, Gromowski GD, Barrett AD, Fremont DH, Diamond MS. Type- and subcomplex-specific neutralizing antibodies against domain III of dengue virus type 2 envelope protein recognize adjacent epitopes. J Virol 2007; 81:12816-26. [PMID: 17881453 PMCID: PMC2169112 DOI: 10.1128/jvi.00432-07] [Citation(s) in RCA: 215] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neutralization of flaviviruses in vivo correlates with the development of an antibody response against the viral envelope (E) protein. Previous studies demonstrated that monoclonal antibodies (MAbs) against an epitope on the lateral ridge of domain III (DIII) of the West Nile virus (WNV) E protein strongly protect against infection in animals. Based on X-ray crystallography and sequence analysis, an analogous type-specific neutralizing epitope for individual serotypes of the related flavivirus dengue virus (DENV) was hypothesized. Using yeast surface display of DIII variants, we defined contact residues of a panel of type-specific, subcomplex-specific, and cross-reactive MAbs that recognize DIII of DENV type 2 (DENV-2) and have different neutralizing potentials. Type-specific MAbs with neutralizing activity against DENV-2 localized to a sequence-unique epitope on the lateral ridge of DIII, centered at the FG loop near residues E383 and P384, analogous in position to that observed with WNV-specific strongly neutralizing MAbs. Subcomplex-specific MAbs that bound some but not all DENV serotypes and neutralized DENV-2 infection recognized an adjacent epitope centered on the connecting A strand of DIII at residues K305, K307, and K310. In contrast, several MAbs that had poor neutralizing activity against DENV-2 and cross-reacted with all DENV serotypes and other flaviviruses recognized an epitope with residues in the AB loop of DIII, a conserved region that is predicted to have limited accessibility on the mature virion. Overall, our experiments define adjacent and structurally distinct epitopes on DIII of DENV-2 which elicit type-specific, subcomplex-specific, and cross-reactive antibodies with different neutralizing potentials.
Collapse
Affiliation(s)
- Soila Sukupolvi-Petty
- Department of Medicine, Washington University School of Medicine, 660 S. Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Ishikawa T, Takasaki T, Kurane I, Nukuzuma S, Kondo T, Konishi E. Co-immunization with West Nile DNA and inactivated vaccines provides synergistic increases in their immunogenicities in mice. Microbes Infect 2007; 9:1089-95. [PMID: 17644390 DOI: 10.1016/j.micinf.2007.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2006] [Revised: 04/28/2007] [Accepted: 05/03/2007] [Indexed: 11/26/2022]
Abstract
West Nile virus is now distributed throughout many temperate, subtropical and tropical areas: vaccines need to be developed that are affordable for developed and developing countries. Here, we constructed and evaluated a DNA vaccine expressing the premembrane and envelope proteins of West Nile virus (pcWNME). Mice immunized twice with 100 or 10 microg of pcWNME developed high or moderate levels of neutralizing antibodies, respectively. These mice were protected from viremia and death after lethal challenge. Mice immunized with a mixture of 1 microg of pcWNME and a small amount (1/10 dose) of a commercial inactivated vaccine developed moderate levels of neutralizing antibodies, whereas immunization with pcWNME or the inactivated vaccine alone induced only low or undetectable levels: co-immunization with the DNA and protein vaccines synergistically increased their own immunogenicities. The synergism reduced the amount of DNA sufficient to induce neutralizing antibodies: a single immunization with doses as low as 0.1 microg induced a titer of 1:40 at a 90% plaque reduction 6 or 9 weeks after immunization. Both IgG1 and IgG2a antibodies were induced in mice by co-immunization with the DNA and protein vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Female
- Immunoglobulin G/immunology
- Injections, Jet
- Mice
- Mice, Inbred ICR
- Vaccines, Combined/immunology
- Vaccines, Combined/pharmacology
- Vaccines, DNA/immunology
- Vaccines, DNA/pharmacology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/pharmacology
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- West Nile Virus Vaccines/immunology
- West Nile Virus Vaccines/pharmacology
- West Nile virus/immunology
Collapse
Affiliation(s)
- Tomohiro Ishikawa
- Department of Health Sciences, Kobe University School of Medicine, 7-10-2 Tomogaoka, Suma-ku, Kobe 654-0142, Japan
| | | | | | | | | | | |
Collapse
|
34
|
Goncalvez AP, Engle RE, St. Claire M, Purcell RH, Lai CJ. Monoclonal antibody-mediated enhancement of dengue virus infection in vitro and in vivo and strategies for prevention. Proc Natl Acad Sci U S A 2007; 104:9422-7. [PMID: 17517625 PMCID: PMC1868655 DOI: 10.1073/pnas.0703498104] [Citation(s) in RCA: 286] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Infection with dengue virus (DENV) or any other flavivirus induces cross-reactive, but weakly neutralizing or nonneutralizing, antibodies that recognize epitopes involving the fusion peptide in the envelope glycoprotein. Humanized mAb IgG 1A5, derived from a chimpanzee, shares properties of cross-reactive antibodies. mAb IgG 1A5 up-regulated DENV infection by a mechanism of antibody-dependent enhancement (ADE) in a variety of Fc receptor-bearing cells in vitro. A 10- to 1,000-fold increase of viral yield in K562 cells, dependent on the DENV serotype, was observed over a range of subneutralizing concentrations of IgG 1A5. A significant increase of DENV-4 viremia titers (up to 100-fold) was also demonstrated in juvenile rhesus monkeys immunized with passively transferred dilutions of IgG 1A5. These results, together with earlier findings of ADE of DENV-2 infection by a polyclonal serum, establish the primate model for analysis of ADE. Considering the abundance of these cross-reactive antibodies, our observations confirm that significant viral amplification could occur during DENV infections in humans with prior infection or with maternally transferred immunity, possibly leading to severe dengue. Strategies to eliminate ADE were explored by altering the antibody Fc structures responsible for binding to Fc receptors. IgG 1A5 variants, containing amino acid substitutions from the Fc region of IgG2 or IgG4 antibodies, reduced but did not eliminate DENV-4-enhancing activity in K562 cells. Importantly, a 9-aa deletion at the N terminus of the CH(2) domain in the Fc region abrogated the enhancing activity.
Collapse
Affiliation(s)
- Ana P. Goncalvez
- *Molecular Viral Biology Section and
- To whom correspondence may be addressed. E-mail: , , or
| | - Ronald E. Engle
- Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | | | - Robert H. Purcell
- Hepatitis Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
- To whom correspondence may be addressed. E-mail: , , or
| | - Ching-Juh Lai
- *Molecular Viral Biology Section and
- To whom correspondence may be addressed. E-mail: , , or
| |
Collapse
|
35
|
Kulkarni-Kale U, Bhosle SG, Manjari GS, Joshi M, Bansode S, Kolaskar AS. Curation of viral genomes: challenges, applications and the way forward. BMC Bioinformatics 2006; 7 Suppl 5:S12. [PMID: 17254296 PMCID: PMC1764468 DOI: 10.1186/1471-2105-7-s5-s12] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Whole genome sequence data is a step towards generating the 'parts list' of life to understand the underlying principles of Biocomplexity. Genome sequencing initiatives of human and model organisms are targeted efforts towards understanding principles of evolution with an application envisaged to improve human health. These efforts culminated in the development of dedicated resources. Whereas a large number of viral genomes have been sequenced by groups or individuals with an interest to study antigenic variation amongst strains and species. These independent efforts enabled viruses to attain the status of 'best-represented taxa' with the highest number of genomes. However, due to lack of concerted efforts, viral genomic sequences merely remained as entries in the public repositories until recently. RESULTS VirGen is a curated resource of viral genomes and their analyses. Since its first release, it has grown both in terms of coverage of viral families and development of new modules for annotation and analysis. The current release (2.0) includes data for twenty-five families with broad host range as against eight in the first release. The taxonomic description of viruses in VirGen is in accordance with the ICTV nomenclature. A well-characterised strain is identified as a 'representative entry' for every viral species. This non-redundant dataset is used for subsequent annotation and analyses using sequenced-based Bioinformatics approaches. VirGen archives precomputed data on genome and proteome comparisons. A new data module that provides structures of viral proteins available in PDB has been incorporated recently. One of the unique features of VirGen is predicted conformational and sequential epitopes of known antigenic proteins using in-house developed algorithms, a step towards reverse vaccinology. CONCLUSION Structured organization of genomic data facilitates use of data mining tools, which provides opportunities for knowledge discovery. One of the approaches to achieve this goal is to carry out functional annotations using comparative genomics. VirGen, a comprehensive viral genome resource that serves as an annotation and analysis pipeline has been developed for the curation of public domain viral genome data http://bioinfo.ernet.in/virgen/virgen.html. Various steps in the curation and annotation of the genomic data and applications of the value-added derived data are substantiated with case studies.
Collapse
Affiliation(s)
| | | | | | - Manali Joshi
- Bioinformatics Centre, University of Pune, Pune 411 007 India
| | - Sandeep Bansode
- Bioinformatics Centre, University of Pune, Pune 411 007 India
| | | |
Collapse
|
36
|
Stiasny K, Kiermayr S, Holzmann H, Heinz FX. Cryptic properties of a cluster of dominant flavivirus cross-reactive antigenic sites. J Virol 2006; 80:9557-68. [PMID: 16973559 PMCID: PMC1617264 DOI: 10.1128/jvi.00080-06] [Citation(s) in RCA: 183] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
A number of flaviviruses are important human pathogens, including yellow fever, dengue, West Nile, Japanese encephalitis, and tick-borne encephalitis (TBE) viruses. Infection with or immunization against any of these viruses induces a subset of antibodies that are broadly flavivirus cross-reactive but do not exhibit significant cross-neutralization. Nevertheless, these antibodies can efficiently bind to the major envelope protein (E), which is the main target of neutralizing and protective antibodies because of its receptor-binding and membrane fusion functions. The structural basis for this phenomenon is still unclear. In our studies with TBE virus, we have provided evidence that such cross-reactive antibodies are specific for a cluster of epitopes that are partially occluded in the cage-like assembly of E proteins at the surfaces of infectious virions and involve-but are not restricted to-amino acids of the highly conserved internal fusion peptide loop. Virus disintegration leads to increased accessibility of these epitopes, allowing the cross-reactive antibodies to bind with strongly increased avidity. The cryptic properties of these sites in the context of infectious virions can thus provide an explanation for the observed lack of efficient neutralizing activity of broadly cross-reactive antibodies, despite their specificity for a functionally important structural element in the E protein.
Collapse
Affiliation(s)
- Karin Stiasny
- Institute of Virology, Medical University of Vienna, Kinderspitalgasse 15, A-1095 Vienna, Austria
| | | | | | | |
Collapse
|
37
|
Development of an effective Japanese encephalitis virus-specific DNA vaccine. Microbes Infect 2006; 8:2578-86. [PMID: 16949850 DOI: 10.1016/j.micinf.2006.06.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 05/24/2006] [Accepted: 06/05/2006] [Indexed: 11/27/2022]
Abstract
Intramuscular immunization with DNA vaccines has been shown to induce a broad range of immune responses and protective immunity in many animal models, but it is less effective in primates. One reason for this may be the low expression of vector-encoded antigen in cells. Here we report that the use of vaccine vector (pCJ-3) containing two regulatory elements, a chimeric intron and a bovine growth hormone (BGH) polyadenylation signal, markedly increased antigen expression both in vitro and in vivo. A positive correlation was seen between the level of expression of Japanese encephalitis virus (JEV) envelope proteins and the levels of antibodies in C3H/HeN mice. Immunization of mice with pCJ-3/ME (pCJ-3 containing the entire membrane and envelope protein genes) with or without cardiotoxin pretreatment resulted in higher antibody titers than immunization with vector containing only envelope protein and conferred full protection against infection with JEV. Electron microscopy showed that pCJ-3/ME expression resulted in the production of virus-like particles of JEV in vitro. The particles enhanced the production of higher titers of neutralizing antibodies and thus provided immunity against JEV. Consequently, the efficacy of the newly developed DNA vaccines was validated. This should pave the way to clinical trials in man.
Collapse
|
38
|
Lim CS, Chua JJE, Wilkerson J, Chow VTK. Differential dengue cross-reactive and neutralizing antibody responses in BALB/c and Swiss albino mice induced by immunization with flaviviral vaccines and by infection with homotypic dengue-2 virus strains. Viral Immunol 2006; 19:33-41. [PMID: 16553548 DOI: 10.1089/vim.2006.19.33] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We investigated whether cross-reactive and/or cross-protective antibodies against dengue virus could be generated in 6-week-old BALB/c mice by immunization with currently approved flaviviral vaccines, i.e., Japanese encephalitis (JE) BIKEN and yellow fever (YF) 17D. Cross-reactivity with dengue antigens was apparent in at least one-third each of JE-vaccinated mouse sera and of JE/YF-vaccinated mouse sera by dengue enzyme immunoassay, but was not detected in sera of mice immunized with YF vaccine alone. All the immunized BALB/c mice failed to generate neutralizing antibodies against the New Guinea C laboratory (NGC-lab) strain of dengue virus type 2. In addition, we determined the specificity of neutralizing antibodies elicited in 3-week-old Swiss albino mice against two homotypic dengue-2 strains, i.e., NGC-lab and Singapore 1999 (SING/99). Although sera from virus-inoculated mice displayed better neutralization against the corresponding strain, antibodies elicited by NGC-lab exhibited a significantly poorer neutralizing response against the SING/99 strain compared to antibodies elicited by SING/99 against NGC-lab. The differences may be related to sequence variations of approximately 3% between the envelope proteins of both strains. Amino acid disparities at positions 71 (Glu --> Ala), 112 (Ser --> Gly) and 124 (Ile --> Asn), which are found in dengue-2 neutralization escape mutants, were also found in the SING/99 strain. The envelope sequence differences may explain diminished binding of NGC-lab-induced neutralizing antibodies to neutralizing epitopes within the envelope of the SING/99 strain, resulting in a lower titer of neutralizing antibodies against another strain of the same serotype.
Collapse
Affiliation(s)
- C S Lim
- Programme in Infectious Diseases, Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
39
|
Marriott L, Willoughby K, Chianini F, Dagleish MP, Scholes S, Robinson AC, Gould EA, Nettleton PF. Detection of Louping ill virus in clinical specimens from mammals and birds using TaqMan RT-PCR. J Virol Methods 2006; 137:21-8. [PMID: 16814876 DOI: 10.1016/j.jviromet.2006.05.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2006] [Revised: 05/18/2006] [Accepted: 05/25/2006] [Indexed: 11/19/2022]
Abstract
The identification of Louping ill virus (LIV) in clinical specimens has been routinely achieved by virus isolation using susceptible pig kidney cells and subsequent serological analysis. While this method is sensitive and detects infectious virus, it is relatively labour intensive and time-consuming. In view of the veterinary and potential medical importance of LIV, a rapid and precise detection method for routine use that employs the TaqMan reverse transcription polymerase chain reaction (RT-PCR) has been developed to detect LIV RNA extracted from field samples. The TaqMan assay was evaluated against virus isolation using 22 cell culture grown LIV isolates, which had previously been partially characterised by sequencing, and material from 63 suspect field cases. Histopathological and/or serological reports were available for 39 of the suspect cases, providing additional diagnostic information to evaluate the results obtained from the TaqMan RT-PCR assay. The TaqMan assay was as sensitive as the cell culture infectious virus assay currently used and had the advantage that it was able to detect LIV in clinical specimens from which infectious virus could not be isolated possibly due to the presence of high levels of LIV antibody.
Collapse
Affiliation(s)
- L Marriott
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian EH26 0PZ, UK.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Li J, Bhuvanakantham R, Howe J, Ng ML. The glycosylation site in the envelope protein of West Nile virus (Sarafend) plays an important role in replication and maturation processes. J Gen Virol 2006; 87:613-622. [PMID: 16476982 DOI: 10.1099/vir.0.81320-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The complete genome of West Nile (Sarafend) virus [WN(S)V] was sequenced. Phylogenetic trees utilizing the complete genomic sequence, capsid gene, envelope gene and NS5 gene/3' untranslated region of WN(S)V classified WN(S)V as a lineage II virus. A full-length infectious clone of WN(S)V with a point mutation in the glycosylation site of the envelope protein (pWNS-S154A) was constructed. Both growth kinetics and the mode of maturation were affected by this mutation. The titre of the pWNS-S154A virus was lower than the wild-type virus. This defect was corrected by the expression of wild-type envelope protein in trans. The pWNS-S154A virus matured intracellularly instead of at the plasma membrane as shown for the parental WN(S)V.
Collapse
Affiliation(s)
- J Li
- Flavivirology Laboratory, Department of Microbiology, 5 Science Drive 2, National University of Singapore, Singapore 117597
| | - R Bhuvanakantham
- Flavivirology Laboratory, Department of Microbiology, 5 Science Drive 2, National University of Singapore, Singapore 117597
| | - J Howe
- Flavivirology Laboratory, Department of Microbiology, 5 Science Drive 2, National University of Singapore, Singapore 117597
| | - M-L Ng
- Flavivirology Laboratory, Department of Microbiology, 5 Science Drive 2, National University of Singapore, Singapore 117597
| |
Collapse
|
41
|
Gaunt MW, Gould EA. Rapid subgroup identification of the flaviviruses using degenerate primer E-gene RT-PCR and site specific restriction enzyme analysis. J Virol Methods 2005; 128:113-27. [PMID: 15927275 DOI: 10.1016/j.jviromet.2005.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2004] [Revised: 04/19/2005] [Accepted: 04/20/2005] [Indexed: 10/25/2022]
Abstract
A simplified and rapid method for the diagnosis of all flaviviruses could provide an important tool for understanding their epidemiology. A protocol based on the use of degenerate nested oligonucleotide primers and RT-PCR was developed for the identification of flaviviruses. The primers were selected to flank the three E-gene markers that identify the viruses, giving DNA products of 971-986 (outer primers) and 859-884 bp (inner primers). Eighty five percent of E genes from flaviviruses representing most of the genus were specifically amplified, representing viruses from each of the 14 virus groups defined by the seventh International Committee for the Taxonomy of Viruses. Categorisation of the flavivirus cDNA products into the corresponding virus groups was undertaken through restriction enzyme analysis by defining conserved restriction sites common to related viruses in appropriate virus groups. Ninety percent of the known vector-borne flaviviruses with published full length E-gene sequences could be identified within 10 h.
Collapse
Affiliation(s)
- M W Gaunt
- Pathogen Molecular Biology and Biochemistry Unit, Department of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| | | |
Collapse
|
42
|
Zhao Z, Date T, Li Y, Kato T, Miyamoto M, Yasui K, Wakita T. Characterization of the E-138 (Glu/Lys) mutation in Japanese encephalitis virus by using a stable, full-length, infectious cDNA clone. J Gen Virol 2005; 86:2209-2220. [PMID: 16033968 DOI: 10.1099/vir.0.80638-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
A stable plasmid DNA, pMWJEAT, was constructed by using full-length Japanese encephalitis virus (JEV) cDNA isolated from the wild-type strain JEV AT31. Recombinant JEV was obtained by synthetic RNA transfection into Vero cells and designated rAT virus. JEV rAT exhibited similar large-plaque morphology and antigenicity to the parental AT31 strain. Mutant clone pMWJEAT-E138K, containing a single Glu-to-Lys mutation at aa 138 of the envelope (E) protein, was also constructed to analyse the mechanisms of viral attenuation arising from this mutation. Recombinant JEV rAT-E138K was also recovered and displayed a smaller-plaque morphology and lower neurovirulence and neuroinvasiveness than either AT31 virus or rAT virus. JEV rAT-E138K exhibited greater plaque formation than rAT virus in virus-cell interactions under acidic conditions. Heparin or heparinase III treatment inhibited binding to Vero cells more efficiently for JEV rAT-E138K than for rAT virus. Inhibition of virus-cell interactions by using wheatgerm agglutinin was more effective for JEV rAT than for rAT-E138K on Vero cells. About 20 % of macropinoendocytosis of JEV rAT for Vero cells was inhibited by cytochalasin D treatment, but no such inhibition occurred for rAT-E138K virus. Furthermore, JEV rAT was predominantly secreted from infected cells, whereas rAT-E138K was more likely to be retained in infected cells. This study demonstrates clearly that a single Glu-to-Lys mutation at aa 138 of the envelope protein affects multiple steps of the viral life cycle. These multiple changes may induce substantial attenuation of JEV.
Collapse
Affiliation(s)
- Zijiang Zhao
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| | - Tomoko Date
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| | - Yuhua Li
- Chengdu Institute of Biological Products, Chengdu 610063, Sichuan Province, PR China
| | - Takanobu Kato
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| | - Michiko Miyamoto
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| | - Kotaro Yasui
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| | - Takaji Wakita
- Department of Microbiology, Tokyo Metropolitan Institute for Neuroscience, 2-6 Musashidai, Fuchu-shi, Tokyo 183-8526, Japan
| |
Collapse
|
43
|
Vlaycheva L, Nickells M, Droll DA, Chambers TJ. Neuroblastoma cell-adapted yellow fever virus: mutagenesis of the E protein locus involved in persistent infection and its effects on virus penetration and spread. J Gen Virol 2005; 86:413-421. [PMID: 15659761 DOI: 10.1099/vir.0.80314-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Persistent infection of mouse neuroblastoma NB41A3 cells with yellow fever 17D virus generates viral variants which exhibit defective cell penetration, poor cell-to-cell spread, small plaque size and reduced growth efficiency, caused by substitution of glycine for aspartic acid or glutamic acid at positions 360 and 362 in the envelope protein. These positions occur within a charge cluster, Asp360-Asp361-Glu362, located in domain III, near its interface with domain I. To characterize further the molecular basis for the variant phenotype, a series of mutant viruses containing substitutions at position 360, 361 and 362, were studied for effects on the cell culture properties typical of the neuroblastoma-adapted variant. Most substitutions at position 360 gave rise to viruses that were very defective in cell penetration, growth efficiency and cell-to-cell spread, whereas substitution with glutamic acid yielded a virus indistinguishable from parental yellow fever 17D. Substitution with lysine was not tolerated and substitution with asparagine resulted in frequent wild-type revertants. A glycine residue was not tolerated at position 361, but substitution at 362 yielded a small plaque virus, similar to the effect of substitution at position 360. These data indicate that the yellow fever virus E protein contains a locus within domain III where a negative-charge cluster is important for optimal function of this domain in virus-cell interactions beyond the stage of virus attachment. Modelling predictions suggest that the mutations alter the local properties of the loop within domain III, and may compromise interactions of this domain with an adjacent region of domain I during conformational changes that occur in the E protein in association with virus entry.
Collapse
Affiliation(s)
- Leonssia Vlaycheva
- Department of Molecular Microbiology and Immunology, St Louis University Health Sciences Center, 1402 South Grand Avenue, St Louis, MO 63104, USA
| | - Michael Nickells
- Department of Molecular Microbiology and Immunology, St Louis University Health Sciences Center, 1402 South Grand Avenue, St Louis, MO 63104, USA
| | - Deborah A Droll
- Department of Molecular Microbiology and Immunology, St Louis University Health Sciences Center, 1402 South Grand Avenue, St Louis, MO 63104, USA
| | - Thomas J Chambers
- Department of Molecular Microbiology and Immunology, St Louis University Health Sciences Center, 1402 South Grand Avenue, St Louis, MO 63104, USA
| |
Collapse
|
44
|
Goncalvez AP, Purcell RH, Lai CJ. Epitope determinants of a chimpanzee Fab antibody that efficiently cross-neutralizes dengue type 1 and type 2 viruses map to inside and in close proximity to fusion loop of the dengue type 2 virus envelope glycoprotein. J Virol 2004; 78:12919-28. [PMID: 15542644 PMCID: PMC525008 DOI: 10.1128/jvi.78.23.12919-12928.2004] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The epitope determinants of chimpanzee Fab antibody 1A5, which have been shown to be broadly reactive to flaviviruses and efficient for cross-neutralization of dengue virus type 1 and type 2 (DENV-1 and DENV-2), were studied by analysis of DENV-2 antigenic variants. Sequence analysis showed that one antigenic variant contained a Gly-to-Val substitution at position 106 within the flavivirus-conserved fusion peptide loop of the envelope protein (E), and another variant contained a His-to-Gln substitution at position 317 in E. Substitution of Gly(106)Val in DENV-2 E reduced the binding affinity of Fab 1A5 by approximately 80-fold, whereas substitution of His(317)Gln had little or no effect on antibody binding compared to the parental virus. Treatment of DENV-2 with beta-mercaptoethanol abolished binding of Fab 1A5, indicating that disulfide bridges were required for the structural integrity of the Fab 1A5 epitope. Binding of Fab 1A5 to DENV-2 was competed by an oligopeptide containing the fusion peptide sequence as shown by competition enzyme-linked immunosorbent assay. Both DENV-2 antigenic variants were shown to be attenuated, or at least similar to the parental virus, when evaluated for growth in cultured cells or for neurovirulence in mice. Fab 1A5 inhibited low pH-induced membrane fusion of mosquito C6/36 cells infected with DENV-1 or DENV-2, as detected by reduced syncytium formation. Both substitutions in DENV-2 E lowered the pH threshold for membrane fusion, as measured in a fusion-from-within assay. In the three-dimensional structure of E, Gly(106) in domain II and His(317) in domain III of the opposite E monomer were spatially close. From the locations of these amino acids, Fab 1A5 appears to recognize a novel epitope that has not been mapped before with a flavivirus monoclonal antibody.
Collapse
Affiliation(s)
- Ana P Goncalvez
- Molecular Viral Biology Section, Laboratory of Infectious Diseases, NIAID, NIH, Building 50, Room 6349, 50 South Dr., MSC 8009, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
45
|
Men R, Yamashiro T, Goncalvez AP, Wernly C, Schofield DJ, Emerson SU, Purcell RH, Lai CJ. Identification of chimpanzee Fab fragments by repertoire cloning and production of a full-length humanized immunoglobulin G1 antibody that is highly efficient for neutralization of dengue type 4 virus. J Virol 2004; 78:4665-74. [PMID: 15078949 PMCID: PMC387713 DOI: 10.1128/jvi.78.9.4665-4674.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A safe and effective dengue vaccine is still not available. Passive immunization with monoclonal antibodies from humans or nonhuman primates represents an attractive alternative for the prevention of dengue virus infection. Fab monoclonal antibodies to dengue type 4 virus (DENV-4) were recovered by repertoire cloning of bone marrow mRNAs from an immune chimpanzee and analyzed for antigen binding specificity, V(H) and V(L) sequences, and neutralizing activity against DENV-4 in vitro. Fabs 5A7, 3C1, 3E4, and 7G4 were isolated from a library constructed from a chimpanzee following intrahepatic transfection with infectious DENV-4 RNA. Fabs 5H2 and 5D9, which had nearly identical V(H) sequences but varied in their V(L) sequences, were recovered from a library constructed from the same chimpanzee after superinfection with a mixture of DENV-1, DENV-2, and DENV-3. In radioimmunoprecipitation, Fab 5A7 precipitated only DENV-4 prM, and Fabs 3E4, 7G4, 5D9, and 5H2 precipitated DENV-4 E but little or no prM. Fab 3E4 and Fab 7G4 competed with each other for binding to DENV-4 in an enzyme-linked immunosorbent assay, as did Fab 3C1 and Fab 5A7. Fab 5H2 recognized an epitope on DENV-4 that was separate from the epitope(s) recognized by other Fabs. Both Fab 5H2 and Fab 5D9 neutralized DENV-4 efficiently with a titer of 0.24 to 0.58 micro g/ml by plaque reduction neutralization test (PRNT), whereas DENV-4-neutralizing activity of other Fabs was low or not detected. Fab 5H2 was converted to full-length immunoglobulin G1 (IgG1) by combining it with human sequences. The humanized chimpanzee antibody IgG1 5H2 produced in CHO cells neutralized DENV-4 strains from different geographical origins at a similar 50% plaque reduction (PRNT(50)) titer of 0.03 to 0.05 micro g/ml. The DENV-4 binding affinities were 0.42 nM for Fab 5H2 and 0.24 nM for full-length IgG1 5H2. Monoclonal antibody IgG1 5H2 may prove valuable for passive immunoprophylaxis against dengue virus in humans.
Collapse
Affiliation(s)
- Ruhe Men
- Molecular Viral Biology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Jääskeläinen A, Han X, Niedrig M, Vaheri A, Vapalahti O. Diagnosis of tick-borne encephalitis by a mu-capture immunoglobulin M-enzyme immunoassay based on secreted recombinant antigen produced in insect cells. J Clin Microbiol 2003; 41:4336-42. [PMID: 12958266 PMCID: PMC193853 DOI: 10.1128/jcm.41.9.4336-4342.2003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Acute tick-borne encephalitis is diagnosed by detection of IgM antibodies to tick-borne encephalitis virus (TBEV) (genus Flavivirus) in patient serum. TBEV membrane (M) and envelope (E) proteins have previously been shown to form virus-like particles when expressed in mammalian cells. We expressed the prM/M and E proteins in insect cells with a recombinant baculovirus system and obtained antigenic protein secreted into the cell culture medium, as evidenced by detection by a panel of five monoclonal antibodies to TBEV E protein. According to the sedimentation pattern in sucrose gradient centrifugation, the proteins were most likely secreted as virus-like particles. A mu-capture immunoglobulin M-enzyme immunoassay (IgM-EIA) test was developed and compared to a commercially available TBEV-IgM test (Progen) based on inactivated purified TBEVs. With a panel of 100 TBEV-IgM-negative, 50 TBEV-IgM-positive, and seven dengue virus-IgM-positive serum samples from our diagnostic laboratory, a sensitivity of 100% and specificity of 99% were obtained, and the correlation coefficient of EIA absorbances with the reference test was 0.93. The antigen was also suitable for IgG antibody detection in an immunofluorescent assay format. This is the first time that secreted, fully antigenic E protein has been produced in insect cells for this arthropod-borne flavivirus.
Collapse
Affiliation(s)
- Anu Jääskeläinen
- Department of Virology, Haartman Institute, Faculty of Veterinary Medicine, FIN-00014 University of Helsinki, Finland.
| | | | | | | | | |
Collapse
|
47
|
Gomez I, Marx F, Saurwein-Teissl M, Gould EA, Grubeck-Loebenstein B. Characterization of Tick-Borne Encephalitis Virus–Specific Human T Lymphocyte Responses by Stimulation with Structural TBEV Proteins Expressed in a Recombinant Baculovirus. Viral Immunol 2003; 16:407-14. [PMID: 14583154 DOI: 10.1089/088282403322396190] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Very little information is available on human T cell responses following exposure to tick-borne encephalitis virus (TBEV) proteins, largely because the virus is a dangerous pathogen and relatively large amounts of purified antigen would be required for the functional characterization of cellular immune responses. We have produced recombinant TBEV proteins using the baculovirus expression system and tested them for their capacity to stimulate T cells in vitro. T lymphocytes from TBEV vaccinated individuals were characterized. The recombinant E and C proteins triggered CD4+ but not CD8+ cells to proliferate and to produce IFN-gamma and IL-5. T cell responses against recombinant NS3 protein were not detected. T cell lines with specificity for the E protein were also established. These lines were CD4+ and had a TH0 cytokine production pattern. Our results demonstrate the utility of recombinant viral proteins to study the generation and characterization of TBEV specific T cell responses.
Collapse
Affiliation(s)
- I Gomez
- Institute for Biomedical Ageing Research, Austrian Academy of Sciences, Innsbruck, Austria
| | | | | | | | | |
Collapse
|
48
|
Zulueta A, Hermida L, Lazo L, Valdés I, Rodríguez R, López C, Silva R, Rosario D, Martín J, Guzmán MG, Guillén G. The fusion site of envelope fragments from each serotype of Dengue virus in the P64k protein, influence some parameters of the resulting chimeric constructs. Biochem Biophys Res Commun 2003; 308:619-26. [PMID: 12914796 DOI: 10.1016/s0006-291x(03)01411-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To characterize the effect of the envelope fragment fusion site in the P64k protein from Neisseria meningitidis several chimeric constructs were obtained. One variant consisted in the insertion of the E fragment from each Dengue serotype within the lipoil binding domain of the P64k, whereas the other was based on the fusion of the envelope fragment at the C-terminus of the same meningoccocal protein. The results of the expression study revealed the majoritary levels with the C-terminus fusion variants of each serotype. In contrast, the highest proportion of soluble protein was reached with the insertion variants independently of the viral serotype. On the other hand, a significant level of degradation was detected for the semipurified forms of the insertion variants being remarkable in the Dengue 2 construct. Finally, the recognition by Dengue murine antibodies was similar independently of the fusion site. Regarding these results, we can affirm the suitability of the C-terminus fusion variants for further vaccine development as well as for a diagnostic system.
Collapse
Affiliation(s)
- Aída Zulueta
- División de Vacunas, Centro de Ingeniería Genética y Biotecnologi;a, Habana, Cuba.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kojima A, Yasuda A, Asanuma H, Ishikawa T, Takamizawa A, Yasui K, Kurata T. Stable high-producer cell clone expressing virus-like particles of the Japanese encephalitis virus e protein for a second-generation subunit vaccine. J Virol 2003; 77:8745-55. [PMID: 12885894 PMCID: PMC167253 DOI: 10.1128/jvi.77.16.8745-8755.2003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We produced and characterized a cell clone (J12#26 cells) that stably expresses Japanese encephalitis virus (JEV) cDNA, J12, which encodes the viral signal peptide, premembrane (prM), and envelope (E) proteins (amino acid positions 105 to 794). Rabbit kidney-derived RK13 cells were transfected with a J12 expression plasmid, selected by resistance to marker antibiotics, and cloned by two cycles of a limiting-dilution method in the presence of antibiotics, a procedure that prevents the successful generation of E-producing cell clones. J12#26 cells secreted virus-like particles containing the authentic E antigen (E-VLP) into the culture medium in a huge enzyme-linked immunosorbent assay-equivalent amount (2.5 micro g per 10(4) cells) to the internationally licensed JE vaccine JE-VAX. E-VLP production was stable after multiple cell passages and persisted over 1 year with 100% expressing cells without detectable cell fusion, apoptosis, or cell death, but was suspended when the cells grew to 100% confluency and contact inhibition occurred. Mice immunized with the purified J12#26 E-antigen without adjuvant developed high titers of neutralizing antibodies for at least 7 months and 100% protection against intraperitoneal challenge with 5 x 10(6) PFU of JEV when examined according to the JE vaccine standardization protocol. These results suggest that the recombinant E-VLP antigen produced by the J12#26 cell clone is an effective, safe, and low-cost second-generation subunit JE vaccine.
Collapse
Affiliation(s)
- Asato Kojima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 162-8640, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Seligman SJ, Bucher DJ. The importance of being outer: consequences of the distinction between the outer and inner surfaces of flavivirus glycoprotein E. Trends Microbiol 2003; 11:108-10. [PMID: 12648938 DOI: 10.1016/s0966-842x(03)00005-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Glycoprotein E of West Nile, dengue and other flaviviruses is the principal stimulus for the development of neutralizing antibodies and contains a fusion peptide responsible for inserting the virus into the host cell membrane. This glycoprotein lies flat on the surface of the virion and therefore only epitopes on the outer or lateral surface are important immunogens. Changes in antigen recognition after exposure of the virus to low pH have yielded clues to the fusion process.
Collapse
Affiliation(s)
- Stephen J Seligman
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA.
| | | |
Collapse
|