1
|
Xu Z, Zhang H, Yang D, Wei D, Demongeot J, Zeng Q. The Mathematical Modeling of the Host-Virus Interaction in Dengue Virus Infection: A Quantitative Study. Viruses 2024; 16:216. [PMID: 38399992 PMCID: PMC10891746 DOI: 10.3390/v16020216] [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: 12/01/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Infectious diseases, such as Dengue fever, pose a significant public health threat. Developing a reliable mathematical model plays a crucial role in quantitatively elucidating the kinetic characteristics of antibody-virus interactions. By integrating previous models and incorporating the antibody dynamic theory, we have constructed a novel and robust model that can accurately simulate the dynamics of antibodies and viruses based on a comprehensive understanding of immunology principles. It explicitly formulates the viral clearance effect of antibodies, along with the positive feedback stimulation of virus-antibody complexes on antibody regeneration. In addition to providing quantitative insights into the dynamics of antibodies and viruses, the model exhibits a high degree of accuracy in capturing the kinetics of viruses and antibodies in Dengue fever patients. This model offers a valuable solution to modeling the differences between primary and secondary Dengue infections concerning IgM/IgG antibodies. Furthermore, it demonstrates that a faster removal rate of antibody-virus complexes might lead to a higher peak viral loading and worse clinical symptom. Moreover, it provides a reasonable explanation for the antibody-dependent enhancement of heterogeneous Dengue infections. Ultimately, this model serves as a foundation for constructing an optimal mathematical model to combat various infectious diseases in the future.
Collapse
Affiliation(s)
- Zhaobin Xu
- School of Life Science, Dezhou University, Dezhou 253023, China
| | - Hongmei Zhang
- School of Life Science, Dezhou University, Dezhou 253023, China
| | - Dongying Yang
- School of Medicine, Dezhou University, Dezhou 253023, China
| | - Dongqing Wei
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, China;
| | - Jacques Demongeot
- Laboratory AGEIS EA 7407, Team Tools for e-Gnosis Medical, Faculty of Medicine, University Grenoble Alpes (UGA), 38700 La Tronche, France;
| | - Qiangcheng Zeng
- School of Life Science, Dezhou University, Dezhou 253023, China
| |
Collapse
|
2
|
Mushtaq S, Khan MIU, Khan MT, Lodhi MS, Wei DQ. Novel mutations in structural proteins of dengue virus genomes. J Infect Public Health 2023; 16:1971-1981. [PMID: 37879150 DOI: 10.1016/j.jiph.2023.10.005] [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: 03/16/2023] [Revised: 09/01/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Genomic characterization of the dengue virus (DENV) is useful for understanding its molecular evolution, transmission, pathogenicity and infectivity. The DENV genomic RNA encodes three structural proteins, capsid (C) envelope (E) and membrane (M) proteins mediating viral entry and assembly during host infection. The current study aims to explore the DENV serotypes and mutations in the E and M proteins. METHODS Twenty-three samples of DENV-positive patients were processed and selected for whole genome sequencing (WGS) from the Punjab Province of Pakistan. RESULTS Among the 23 WGS, 19 samples showed numerous mutations (BioProject ID PRJNA943555). DENV1 and DENV2 are the most prevalent serotypes. A total of 179 mutations were detected in the E protein, in which K203E, T88A, I114L, and I293T are novel. The I270L, T272A, S273L, and T277A were found in the "kl" β-hairpin (aa 270-279). The M protein harbors 74 mutations, of which 24 were novel. Three prominent complementary regions in the prM and E protein complex formations include R6, E46, D47, D63, and D65 on 'pr' peptide, and E84, K64, and H244, K247 on E, remain conserved except R6C. To our knowledge, it is the first comprehensive study of mutations in structural proteins. CONCLUSION Genomic epidemiology is critical for analyzing emerging mutations and designing new policies therapeutic efforts for future outbreaks.
Collapse
Affiliation(s)
- Saira Mushtaq
- Institute of Molecular Biology and Biotechnology, The University of Lahore, KM Defence Road, Postal code: 5881, Lahore, Pakistan.
| | - Malik Ihsan Ullah Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, KM Defence Road, Postal code: 5881, Lahore, Pakistan.
| | - Muhammad Tahir Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, KM Defence Road, Postal code: 5881, Lahore, Pakistan; Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan 473006, PR China.
| | - Madeeha Shahzad Lodhi
- Institute of Molecular Biology and Biotechnology, The University of Lahore, KM Defence Road, Postal code: 5881, Lahore, Pakistan.
| | - Dong Qing Wei
- Zhongjing Research and Industrialization Institute of Chinese Medicine, Zhongguancun Scientific Park, Meixi, Nanyang, Henan 473006, PR China; State Key Laboratory of Microbial Metabolism, Shanghai-Islamabad-Belgrade Joint Innovation Center on Antibacterial Resistances, Joint International Research Laboratory of Metabolic & Developmental Sciences and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, PR China; Peng Cheng Laboratory, Shenzhen, Guangdong 518055, PR China.
| |
Collapse
|
3
|
Estofolete CF, Versiani AF, Dourado FS, Milhim BHGA, Pacca CC, Silva GCD, Zini N, dos Santos BF, Gandolfi FA, Mistrão NFB, Garcia PHC, Rocha RS, Gehrke L, Bosch I, Marques RE, Teixeira MM, da Fonseca FG, Vasilakis N, Nogueira ML. Influence of previous Zika virus infection on acute dengue episode. PLoS Negl Trop Dis 2023; 17:e0011710. [PMID: 37943879 PMCID: PMC10662752 DOI: 10.1371/journal.pntd.0011710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 11/21/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The co-circulation of flaviviruses in tropical regions has led to the hypothesis that immunity generated by a previous dengue infection could promote severe disease outcomes in subsequent infections by heterologous serotypes. This study investigated the influence of antibodies generated by previous Zika infection on the clinical outcomes of dengue infection. METHODOLOGY/PRINCIPAL FINDINGS We enrolled 1,043 laboratory confirmed dengue patients and investigated their prior infection to Zika or dengue. Severe forms of dengue disease were more frequent in patients with previous Zika infection, but not in those previously exposed to dengue. CONCLUSIONS/SIGNIFICANCE Our findings suggest that previous Zika infection may represent a risk factor for subsequent severe dengue disease, but we did not find evidence of antibody-dependent enhancement (higher viral titer or pro-inflammatory cytokine overexpression) contributing to exacerbation of the subsequent dengue infection.
Collapse
Affiliation(s)
- Cassia F. Estofolete
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Alice F. Versiani
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
- Department of Pathology, University of Texas Medical Branch; Galveston, Texas, United States of America
| | - Fernanda S. Dourado
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Bruno H. G. A. Milhim
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Carolina C. Pacca
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Gislaine C. D. Silva
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Nathalia Zini
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Barbara F. dos Santos
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Flora A. Gandolfi
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Natalia F. B. Mistrão
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Pedro H. C. Garcia
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Rodrigo S. Rocha
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Cambridge, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School; Boston, Massachusetts, United States of America
| | - Irene Bosch
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Cambridge, Massachusetts, United States of America
| | - Rafael E. Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM); Campinas, Sao Paulo, Brazil
| | - Mauro M. Teixeira
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais; Belo Horizonte, Minas Gerais, Brazil
| | - Flavio G. da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais; Belo Horizonte, Minas Gerais, Brazil
- Centro de Tecnoogia em Vacinas da UFMG, Universidade Federal de Minas Gerais; Belo Horizonte, Minas Gerais, Brazil
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch; Galveston, Texas, United States of America
- Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch; Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch; Galveston, Texas, United States of America
- Center for Tropical Diseases, University of Texas Medical Branch; Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch; Galveston, Texas, United States of America
| | - Maurício L. Nogueira
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP); São José do Rio Preto, Sao Paulo, Brazil
- Department of Pathology, University of Texas Medical Branch; Galveston, Texas, United States of America
| |
Collapse
|
4
|
Medina-Carrasco D, Pupo D, González-Lodeiro LG, García LE, Martin AM, Huerta V. Activity of domain III-specific antibodies in early convalescence: A case study. Virology 2023; 587:109883. [PMID: 37757730 DOI: 10.1016/j.virol.2023.109883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/15/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
The Dengue virus complex (DENV), formed by four serotypes, constitutes the most important arbovirus affecting humans. The structural domain III of their envelope protein (DIII) elicits strongly neutralizing serotype-specific antibodies. Contrasting results have been obtained regarding their role in the serum neutralizing activity of infected patients. We used a DENV immune serum from a secondary infection to examine the impact of characterizing the anti-DIII antibody response after affinity purification with recombinant DIII proteins to eliminate potential interferences from the interactions with human plasma proteins and other anti-DENV antibodies. Total anti-DENV IgG repertoire and anti-DIIIE antibodies were compared in functionality. In early convalescence, reactivity of anti-DIII antibodies is serotype specific and exhibits the strongest reactivity with infecting serotypes. Purification of anti-DIII antibodies emphasizes the reactivity profile as compared to total IgG fraction and serum. Serotype-specificity of the virus neutralization activity correlated with the apparent kD of the binding to recombinant DIIIs.
Collapse
Affiliation(s)
- Danya Medina-Carrasco
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Dianne Pupo
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Luis G González-Lodeiro
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Lisandra E García
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Alejandro M Martin
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba
| | - Vivian Huerta
- Department of Systems Biology, Biomedical Research Direction, Center for Genetic Engineering and Biotechnology (CIGB), Havana, Cuba.
| |
Collapse
|
5
|
Modak A, Mishra SR, Awasthi M, Sreedevi S, Sobha A, Aravind A, Kuppusamy K, Sreekumar E. Higher-temperature-adapted dengue virus serotype 2 strain exhibits enhanced virulence in AG129 mouse model. FASEB J 2023; 37:e23062. [PMID: 37389962 DOI: 10.1096/fj.202300098r] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/13/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
The factors that drive dengue virus (DENV) evolution, and selection of virulent variants are yet not clear. Higher environmental temperature shortens DENV extrinsic incubation period in mosquitoes, increases human transmission, and plays a critical role in outbreak dynamics. In the present study, we looked at the effect of temperature in altering the virus virulence. We found that DENV cultured at a higher temperature in C6/36 mosquito cells was significantly more virulent than the virus grown at a lower temperature. In a mouse model, the virulent strain induced enhanced viremia and aggressive disease with a short course, hemorrhage, severe vascular permeability, and death. Higher inflammatory cytokine response, thrombocytopenia, and severe histopathological changes in vital organs such as heart, liver, and kidney were hallmarks of the disease. Importantly, it required only a few passages for the virus to acquire a quasi-species population harboring virulence-imparting mutations. Whole genome comparison with a lower temperature passaged strain identified key genomic changes in the structural protein-coding regions as well as in the 3'UTR of the viral genome. Our results point out that virulence-enhancing genetic changes could occur in the dengue virus genome under enhanced growth temperature conditions in mosquito cells.
Collapse
Affiliation(s)
- Ayan Modak
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Srishti Rajkumar Mishra
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Mansi Awasthi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Sreeja Sreedevi
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Archana Sobha
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Arya Aravind
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
| | - Krithiga Kuppusamy
- Bioscience Research & Training Centre (BRTC), Kerala Veterinary and Animal Sciences University, Bio360 Life Sciences Park, Thiruvananthapuram, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, India
- Molecular Bioassay Laboratory, Institute of Advanced Virology (IAV), Bio360 Life Sciences Park, Thiruvananthapuram, India
| |
Collapse
|
6
|
Pannala VR, Nguyen HD, Wallqvist A. A stochastic B cell affinity maturation model to characterize mechanisms of protection for tetravalent dengue vaccine constructs. Front Mol Biosci 2023; 10:1100434. [PMID: 37520320 PMCID: PMC10375700 DOI: 10.3389/fmolb.2023.1100434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Dengue annually infects millions of people from a regionally and seasonally varying dengue virus population circulating as four distinct serotypes. Effective protection against dengue infection and disease requires tetravalent vaccine formulations to stimulate a balanced protective immune response to all four serotypes. However, this has been a challenge to achieve, and several clinical trials with different leading vaccine candidates have demonstrated unbalanced replication and interference of interindividual serotype components, leading to low efficacy and enhanced disease severity for dengue-naïve populations. Production of serotype-specific neutralizing antibodies is largely viewed as a correlate of protection against severe dengue disease. However, the underlying mechanisms that lead to these protective immune responses are not clearly elucidated. In this work, using a stochastic model of B cell affinity maturation, we tested different live-attenuated vaccine constructs with varied viral replication rates and contrasted the initiation and progress of adaptive immune responses during tetravalent vaccination and after dengue virus challenge. Comparison of our model simulations across different disease-severity levels suggested that individual production of high levels of serotype-specific antibodies together with a lower cross-reactive antibody are better correlates for protection. Furthermore, evolution of these serotype-specific antibodies was dependent on the percent of viral attenuation in the vaccine, and production of initial B cell and T cell populations pre- and post-secondary dengue infection was crucial in providing protective immunity for dengue-naïve populations. Furthermore, contrasting disease severity with respect to different dengue serotypes, our model simulations showed that tetravalent vaccines fare better against DENV-4 serotype when compared to other serotypes.
Collapse
Affiliation(s)
- Venkat R. Pannala
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Frederick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Hung D. Nguyen
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Frederick, MD, United States
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, United States
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Frederick, MD, United States
| |
Collapse
|
7
|
Vista FES, Tantengco OAG, Dispo MD, Opiso DMS, Badua CLDC, Gerardo JPZ, Perez JRM, Baldo KAT, Chao DY, Dalmacio LMM. Trends in ELISA-Based Flavivirus IgG Serosurveys: A Systematic Review. Trop Med Infect Dis 2023; 8:tropicalmed8040224. [PMID: 37104349 PMCID: PMC10143827 DOI: 10.3390/tropicalmed8040224] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023] Open
Abstract
Flaviviruses include virus species that are major public health threats worldwide. To determine the immunity landscape of these viruses, seroprevalence studies are often performed using IgG ELISA, which is a simple and rapid alternative to the virus neutralization test. In this review, we aim to describe the trends in flavivirus IgG ELISA-based serosurveys. A systematic literature review using six databases was performed to collate cohort and cross-sectional studies performed on the general population. A total of 204 studies were included in this review. The results show that most studies were performed on dengue virus (DENV), whereas Japanese Encephalitis Virus (JEV) was the least studied. For geographic distribution, serosurveys followed known disease prevalence. Temporally, the number of serosurveys increased after outbreaks and epidemics except for JEV, for which studies were performed to demonstrate the effectiveness of vaccination campaigns. Commercial kits were more commonly used than in-house assays for DENV, West Nile Virus (WNV), and Zika virus (ZIKV). Overall, most studies employed an indirect ELISA format, and the choice of antigens varied per virus. This review shows that flavivirus epidemiology is related to the regional and temporal distribution of serosurveys. It also highlights that endemicity, cross-reactivities, and kit availabilities affect assay choice in serosurveys.
Collapse
Affiliation(s)
- Fatima Ericka S Vista
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Ourlad Alzeus G Tantengco
- Department of Physiology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
- Department of Biology, College of Science, De La Salle University, Manila 0922, Philippines
| | - Micah D Dispo
- Department of Epidemiology and Biostatistics, College of Public Health, University of the Philippines Manila, Manila 1000, Philippines
| | - Danna Mae S Opiso
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Christian Luke D C Badua
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - John Patrick Z Gerardo
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Juan Raphael M Perez
- College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Karol Ann T Baldo
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| | - Day-Yu Chao
- Graduate Institute of Microbiology and Public Health, National Chung Hsing University, Taichung 40227, Taiwan
| | - Leslie Michelle M Dalmacio
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila 1000, Philippines
| |
Collapse
|
8
|
Cao L, Yu Z, He H, Guo X, Wei C, Zhang X, Bao J, Li C, Zhou H, Xin J, Nan F. Retrospective investigation of the origin and epidemiology of the dengue outbreak in Yunnan, China from 2017 to 2018. Front Vet Sci 2023; 10:1137392. [PMID: 37124563 PMCID: PMC10132138 DOI: 10.3389/fvets.2023.1137392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023] Open
Abstract
Since 2013, a dengue epidemic has broken out in Yunnan, China and neighboring countries. However, after the COVID-19 pandemic in 2019, the number of dengue cases decreased significantly. In this retrospective study, epidemiological and genetic diversity characterizations of dengue viruses (DENV) isolated in Yunnan between 2017 and 2018 were performed. The results showed that the dengue outbreak in Yunnan from 2017 to 2018 was mainly caused by DENV1 (genotype I and genotype V) and DENV2 (Asia I, Asia II, and Cosmopolitan). Furthermore, correlation analysis indicated a significant positive correlation between the number of imported and local cases (correlation coefficient = 0.936). Multiple sequence alignment and phylogenetic divergence analysis revealed that the local isolates are closely related to the isolates from Myanmar and Laos. Interestingly, recombination analysis found that the DENV1 and DENV2 isolates in this study had widespread intra-serotype recombination. Taken together, the results of the epidemiological investigation imply that the dengue outbreak in Yunnan was primarily due to imported cases. This study provides a new reference for further investigations on the prevalence and molecular epidemiology of DENV in Yunnan, China.
Collapse
Affiliation(s)
- Liang Cao
- College of Laboratory, Jilin Medical University, Jilin, China
- *Correspondence: Liang Cao
| | - Ziping Yu
- Institute of Virology, Wenzhou University, Wenzhou, China
| | - Haiqiang He
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaofang Guo
- Yunnan Institute of Parasitic Diseases, Pu'er, Yunnan, China
| | - Chun Wei
- Yunnan Institute of Parasitic Diseases, Pu'er, Yunnan, China
| | - Xuancheng Zhang
- College of Veterinary Medicine, Jilin University, Changchun, China
| | - Junduo Bao
- Collage of Agriculture, Yanbian University, Yanbian, China
| | - Chenghui Li
- Collage of Agriculture, Yanbian University, Yanbian, China
- Chenghui Li
| | - Hongning Zhou
- Yunnan Institute of Parasitic Diseases, Pu'er, Yunnan, China
- Hongning Zhou
| | - Jialiang Xin
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Jialiang Xin
| | - Fulong Nan
- Department of Special Medicine, School of Basic Medicine, Qingdao University, Qingdao, China
- Fulong Nan
| |
Collapse
|
9
|
Dengue virus infection - a review of pathogenesis, vaccines, diagnosis and therapy. Virus Res 2023; 324:199018. [PMID: 36493993 DOI: 10.1016/j.virusres.2022.199018] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/19/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
The transmission of dengue virus (DENV) from an infected Aedes mosquito to a human, causes illness ranging from mild dengue fever to fatal dengue shock syndrome. The similar conserved structure and sequence among distinct DENV serotypes or different flaviviruses has resulted in the occurrence of cross reaction followed by antibody-dependent enhancement (ADE). Thus far, the vaccine which can provide effective protection against infection by different DENV serotypes remains the biggest hurdle to overcome. Therefore, deep investigation is crucial for the potent and effective therapeutic drugs development. In addition, the cross-reactivity of flaviviruses that leads to false diagnosis in clinical settings could result to delay proper intervention management. Thus, the accurate diagnostic with high specificity and sensitivity is highly required to provide prompt diagnosis in respect to render early treatment for DENV infected individuals. In this review, the recent development of neutralizing antibodies, antiviral agents, and vaccine candidates in therapeutic platform for DENV infection will be discussed. Moreover, the discovery of antigenic cryptic epitopes, principle of molecular mimicry, and application of single-chain or single-domain antibodies towards DENV will also be presented.
Collapse
|
10
|
Ma X, Yuan Z, Yi Z. Identification and characterization of key residues in Zika virus envelope protein for virus assembly and entry. Emerg Microbes Infect 2022; 11:1604-1620. [PMID: 35612559 PMCID: PMC9196690 DOI: 10.1080/22221751.2022.2082888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Zika virus (ZIKV), a family member in the Flavivirus genus, has re-emerged as a global public health concern. The envelope (E) proteins of flaviviruses play a dual role in viral assembly and entry. To identify the key residues of E in virus entry, we generated a ZIKV trans-complemented particle (ZIKVTCP) system, in which a subgenomic reporter replicon was packaged by trans-complementation with expression of CprME. We performed mutagenesis studies of the loop regions that protrude from the surface of the virion in the E ectodomains (DI, DII, DIII). Most mutated ZIKVTCPs exhibited deficient egress. Mutations in DII and in the hinge region of DI and DIII affected prM expression. With a bioorthogonal system, photocrosslinking experiments identified crosslinked intracellular E trimers and demonstrated that egress-deficient mutants in DIII impaired E trimerization. Of these mutants, an E-trimerization-dead mutation D389A that nears the E-E interface between two neighbouring spikes in the immature virion completely abolished viral egress. Several mutations abolished ZIKVTCPs’ entry, without severely affecting viral egress. Further virus binding experiments demonstrated a deficiency of the mutated ZIKVTCPs in virus attachment. Strikingly, synthesized peptide containing residues of two mutants (268-273aa in DII) could bind to host cells and significantly compete for viral attachment and interfere with viral infection, suggesting an important role of these resides in virus entry. Our findings uncovered the requirement for DIII mediated-E trimerization in viral egress, and discovered a key residue group in DII that participates in virus entry.
Collapse
Affiliation(s)
- Xiao Ma
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, People's Republic of China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, People's Republic of China
| | - Zhigang Yi
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, and Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, People's Republic of China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
11
|
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
|
12
|
Thompson D, Guenther B, Manayani D, Mendy J, Smith J, Espinosa DA, Harris E, Alexander J, Vang L, Morello CS. Zika virus-like particle vaccine fusion loop mutation increases production yield but fails to protect AG129 mice against Zika virus challenge. PLoS Negl Trop Dis 2022; 16:e0010588. [PMID: 35793354 PMCID: PMC9292115 DOI: 10.1371/journal.pntd.0010588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 07/18/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus with maternal infection associated with preterm birth, congenital malformations, and fetal death, and adult infection associated with Guillain-Barré syndrome. Recent widespread endemic transmission of ZIKV and the potential for future outbreaks necessitate the development of an effective vaccine. We developed a ZIKV vaccine candidate based on virus-like-particles (VLPs) generated following transfection of mammalian HEK293T cells using a plasmid encoding the pre-membrane/membrane (prM/M) and envelope (E) structural protein genes. VLPs were collected from cell culture supernatant and purified by column chromatography with yields of approximately 1-2mg/L. To promote increased particle yields, a single amino acid change of phenylalanine to alanine was made in the E fusion loop at position 108 (F108A) of the lead VLP vaccine candidate. This mutation resulted in a modest 2-fold increase in F108A VLP production with no detectable prM processing by furin to a mature particle, in contrast to the lead candidate (parent). To evaluate immunogenicity and efficacy, AG129 mice were immunized with a dose titration of either the immature F108A or lead VLP (each alum adjuvanted). The resulting VLP-specific binding antibody (Ab) levels were comparable. However, geometric mean neutralizing Ab (nAb) titers using a recombinant ZIKV reporter were significantly lower with F108A immunization compared to lead. After virus challenge, all lead VLP-immunized groups showed a significant 3- to 4-Log10 reduction in mean ZIKV RNAemia levels compared with control mice immunized only with alum, but the RNAemia reduction of 0.5 Log10 for F108A groups was statistically similar to the control. Successful viral control by the lead VLP candidate following challenge supports further vaccine development for this candidate. Notably, nAb titer levels in the lead, but not F108A, VLP-immunized mice inversely correlated with RNAemia. Further evaluation of sera by an in vitro Ab-dependent enhancement assay demonstrated that the F108A VLP-induced immune sera had a significantly higher capacity to promote ZIKV infection in FcγR-expressing cells. These data indicate that a single amino acid change in the fusion loop resulted in increased VLP yields but that the immature F108A particles were significantly diminished in their capacity to induce nAbs and provide protection against ZIKV challenge. Zika virus (ZIKV) is transmitted by mosquitoes and is a serious health threat due to potential epidemic spread. Infection in adults may lead to Guillain-Barré syndrome, a neurological disorder, or may cause harm to a developing fetus resulting in preterm birth, fetal death, or devastating congenital malformations. There are currently no approved vaccines against ZIKV. We previously developed a lead candidate vaccine based on a virus-like particle (VLP) that was generated in tissue culture. This ZIKV shell is devoid of any viral genetic material. In previous studies, this lead VLP candidate generated neutralizing antibodies (nAbs) that recognized wild-type ZIKV and prevented viral replication in both mice and non-human primates. To increase production of the lead VLP candidate and decrease cost-of-goods, we introduced a single amino acid change, phenylalanine to alanine, in the envelope glycoprotein. This change resulted in a modest increase in VLP yield. However, this single amino acid change resulted in reduced induction of nAbs following immunization and no significant reduction of RNAemia following challenge compared to the lead candidate. The results of this study suggest this investigational vaccine candidate is not suitable for further vaccine development and that ZIKV VLP maturation may have an important role in protection.
Collapse
Affiliation(s)
- Danielle Thompson
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Ben Guenther
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Darly Manayani
- PaxVax Inc., San Diego, California, United States of America
| | - Jason Mendy
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Jonathan Smith
- PaxVax Inc., San Diego, California, United States of America
| | - Diego A. Espinosa
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Jeff Alexander
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
- PaxVax Inc., San Diego, California, United States of America
| | - Lo Vang
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | | |
Collapse
|
13
|
Hung SJ, Tsai HP, Wang YF, Ko WC, Wang JR, Huang SW. Assessment of the Risk of Severe Dengue Using Intrahost Viral Population in Dengue Virus Serotype 2 Patients via Machine Learning. Front Cell Infect Microbiol 2022; 12:831281. [PMID: 35223554 PMCID: PMC8866709 DOI: 10.3389/fcimb.2022.831281] [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: 12/08/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Dengue virus, a positive-sense single-stranded RNA virus, continuously threatens human health. Although several criteria for evaluation of severe dengue have been recently established, the ability to prognose the risk of severe outcomes for dengue patients remains limited. Mutant spectra of RNA viruses, including single nucleotide variants (SNVs) and defective virus genomes (DVGs), contribute to viral virulence and growth. Here, we determine the potency of intrahost viral population in dengue patients with primary infection that progresses into severe dengue. A total of 65 dengue virus serotype 2 infected patients in primary infection including 17 severe cases were enrolled. We utilized deep sequencing to directly define the frequency of SNVs and detection times of DVGs in sera of dengue patients and analyzed their associations with severe dengue. Among the detected SNVs and DVGs, the frequencies of 9 SNVs and the detection time of 1 DVG exhibited statistically significant differences between patients with dengue fever and those with severe dengue. By utilizing the detected frequencies/times of the selected SNVs/DVG as features, the machine learning model showed high average with a value of area under the receiver operating characteristic curve (AUROC, 0.966 ± 0.064). The elevation of the frequency of SNVs at E (nucleotide position 995 and 2216), NS2A (nucleotide position 4105), NS3 (nucleotide position 4536, 4606), and NS5 protein (nucleotide position 7643 and 10067) and the detection times of the selected DVG that had a deletion junction in the E protein region (nucleotide positions of the junction: between 969 and 1022) increased the possibility of dengue patients for severe dengue. In summary, we demonstrated the detected frequencies/times of SNVs/DVG in dengue patients associated with severe disease and successfully utilized them to discriminate severe patients using machine learning algorithm. The identified SNVs and DVGs that are associated with severe dengue will expand our understanding of intrahost viral population in dengue pathogenesis.
Collapse
Affiliation(s)
- Su-Jhen Hung
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | - Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ya-Fang Wang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
- *Correspondence: Sheng-Wen Huang,
| |
Collapse
|
14
|
Nadugala MN, Jeewandara C, Jadi RS, Malavige GN, de Silva AM, Premaratne PH, Goonasekara CL. Natural immunogenic properties of bioinformatically predicted linear B-cell epitopes of dengue envelope and pre-membrane proteins. BMC Immunol 2021; 22:71. [PMID: 34732126 PMCID: PMC8567598 DOI: 10.1186/s12865-021-00462-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
Background The natural antibody responses to B-cell epitopes from dengue structural proteins were assessed using immune sera from people having well-defined past dengue infections with one of the four serotypes. Method Based on an immune-computational analysis previously conducted, nineteen epitopes from the envelope (E) and eight epitopes from pre-membrane (prM), which were more than 50% conserved across all the four DENV serotypes, were selected. Peptides to represent these B-cell epitopes were obtained from commercially available arrays, and were subjected to enzyme linked immunosorbent assay with sera obtained from dengue seropositive healthy volunteers (DENV1 n = 12: DENV2 n = 12: DENV3 n = 12 and DENV4 n = 12), and 10 dengue seronegative healthy volunteers from Sri Lanka. The cut-off value for the positive antibody response was set by taking the mean response of a peptide to the negative sera plus three standard deviations. The peptides (N = 7) showing the broad immune responses were used to generate antibodies in three mice (Balb/c) batches. The mice antisera were then subjected to microneutralization assays against all the four DENV serotypes. An EC50 viral neutralization ≥ 40 times the serum dilution was considered as neutralizing. Results Five of the E-peptide and two prM peptides were recognised by most individuls exposed to infections with each of the four serotypes, showing a serotype cross-reactive broad antibody response. The mice immune sera against the peptides representing the five E protein epitopes neutralized all the four DENV serotypes. Two of these five epitopes are from the Domain II, whereas one of them includes the whole bc-loop region. Conclusion The antibody responses of highly conserved epitopes across the serotypes, were broadly responsive with sera of all four DENV serotypes collected from individuals infected with only one DENV serotype. Weakly conserved epitopes showed rather specific antibody responses dominated by one or few serotypes. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00462-4.
Collapse
Affiliation(s)
- Mahesha N Nadugala
- Faculty of Medicine, General Sir John Kotelawala Defence University, Ratmalana, Sri Lanka
| | - Chandima Jeewandara
- Allergy Immunology and Cell Biology Unit, University of Sri Jayewardanapura, Gangodawila, Sri Lanka
| | - Ramesh S Jadi
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Gathsaurie N Malavige
- Centre for Dengue Research, University of Sri Jayawardanapura, Gangodawila, Sri Lanka
| | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Prasad H Premaratne
- Faculty of Medicine, General Sir John Kotelawala Defence University, Ratmalana, Sri Lanka
| | - Charitha L Goonasekara
- Faculty of Medicine, General Sir John Kotelawala Defence University, Ratmalana, Sri Lanka.
| |
Collapse
|
15
|
Qu S, Wang X, Yang L, Meng R, Feng C, Yang B, Huang J, Li Q, Wang J, Zhang D. Mapping of a unique epitope on domain III of the envelope protein of Tembusu virus. Virus Res 2021; 306:198582. [PMID: 34599934 DOI: 10.1016/j.virusres.2021.198582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
We recently developed a Tembusu virus (TMUV)-specific monoclonal antibody (MAb) 12F11, which was found to recognize a long amino acid sequence between residues 8 and 77 of domain III of the envelope protein (EDIII). Here, the epitope recognized by MAb 12F11 was mapped using alanine substitutions combined with dissociation constant analysis. The findings, and prediction of tertiary structure of TMUV EDIII, showed that the MAb 12F11 epitope contained one critical residue and 13 peripheral residues. Moreover, the antigenic site was shown to span four loops (N-terminal region, AB, BC, and CD) and three β-strands (A, B, and D). The present work contributes to the understanding of antigenic structure of TMUV envelope protein.
Collapse
Affiliation(s)
- Shenghua Qu
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Chonglun Feng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Baolin Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Jingjing Huang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Qiong Li
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Jiaying Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, Haidian district 100193, People's Republic of China.
| |
Collapse
|
16
|
Sebayang AA, Fahlena H, Anam V, Knopoff D, Stollenwerk N, Aguiar M, Soewono E. Modeling Dengue Immune Responses Mediated by Antibodies: A Qualitative Study. BIOLOGY 2021; 10:biology10090941. [PMID: 34571818 PMCID: PMC8464952 DOI: 10.3390/biology10090941] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary With more than one-third of the world population at risk of acquiring the disease, dengue fever is a major public health problem. Caused by four antigenically distinct but related serotypes, disease severity is associated with the immunological status of the individual, seronegative or seropositive, prior to a natural dengue infection. While a primary natural dengue infection is often asymptomatic or mild, individuals experiencing a secondary dengue infection with a heterologous serotype have higher risk of developing the severe form of the disease, linked to the antibody-dependent enhancement (ADE) process. We develop a modeling framework to describe the dengue immune responses mediated by antibodies. Our model framework can describe qualitatively the dynamic of the viral load and antibodies production for scenarios of primary and secondary infections, as found in the empirical immunology literature. Studies such as the one described here serve as a baseline to further model extensions. Future refinements of our framework will be of use to evaluate the impact of imperfect dengue vaccines. Abstract Dengue fever is a viral mosquito-borne infection and a major international public health concern. With 2.5 billion people at risk of acquiring the infection around the world, disease severity is influenced by the immunological status of the individual, seronegative or seropositive, prior to natural infection. Caused by four antigenically related but distinct serotypes, DENV-1 to DENV-4, infection by one serotype confers life-long immunity to that serotype and a period of temporary cross-immunity (TCI) to other serotypes. The clinical response on exposure to a second serotype is complex with the so-called antibody-dependent enhancement (ADE) process, a disease augmentation phenomenon when pre-existing antibodies to previous dengue infection do not neutralize but rather enhance the new infection, used to explain the etiology of severe disease. In this paper, we present a minimalistic mathematical model framework developed to describe qualitatively the dengue immunological response mediated by antibodies. Three models are analyzed and compared: (i) primary dengue infection, (ii) secondary dengue infection with the same (homologous) dengue virus and (iii) secondary dengue infection with a different (heterologous) dengue virus. We explore the features of viral replication, antibody production and infection clearance over time. The model is developed based on body cells and free virus interactions resulting in infected cells activating antibody production. Our mathematical results are qualitatively similar to the ones described in the empiric immunology literature, providing insights into the immunopathogenesis of severe disease. Results presented here are of use for future research directions to evaluate the impact of dengue vaccines.
Collapse
Affiliation(s)
- Afrina Andriani Sebayang
- Department of Mathematics, Institut Teknologi Bandung, Bandung 40132, Indonesia; (A.A.S.); (H.F.)
| | - Hilda Fahlena
- Department of Mathematics, Institut Teknologi Bandung, Bandung 40132, Indonesia; (A.A.S.); (H.F.)
| | - Vizda Anam
- Basque Centre for Applied Mathematics (BCAM), Alameda Mazarredo, 14, 48009 Bilbao, Spain; (V.A.); (D.K.); (N.S.)
| | - Damián Knopoff
- Basque Centre for Applied Mathematics (BCAM), Alameda Mazarredo, 14, 48009 Bilbao, Spain; (V.A.); (D.K.); (N.S.)
| | - Nico Stollenwerk
- Basque Centre for Applied Mathematics (BCAM), Alameda Mazarredo, 14, 48009 Bilbao, Spain; (V.A.); (D.K.); (N.S.)
- Dipartimento di Matematica, Universita degli Studi di Trento, Via Sommarive 14, 38123 Trento, Italy
| | - Maíra Aguiar
- Basque Centre for Applied Mathematics (BCAM), Alameda Mazarredo, 14, 48009 Bilbao, Spain; (V.A.); (D.K.); (N.S.)
- Dipartimento di Matematica, Universita degli Studi di Trento, Via Sommarive 14, 38123 Trento, Italy
- Ikerbasque, Basque Foundation for Science, Euskadi Plaza, 5, 48009 Bilbo, Spain
- Correspondence: (M.A.); (E.S.)
| | - Edy Soewono
- Department of Mathematics, Institut Teknologi Bandung, Bandung 40132, Indonesia; (A.A.S.); (H.F.)
- Center for Mathematical Modeling and Simulation, Institut Teknologi Bandung, Bandung 40132, Indonesia
- Correspondence: (M.A.); (E.S.)
| |
Collapse
|
17
|
Zecchin B, Fusaro A, Milani A, Schivo A, Ravagnan S, Ormelli S, Mavian C, Michelutti A, Toniolo F, Barzon L, Monne I, Capelli G. The central role of Italy in the spatial spread of USUTU virus in Europe. Virus Evol 2021; 7:veab048. [PMID: 34513027 PMCID: PMC8427344 DOI: 10.1093/ve/veab048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
USUTU virus (USUV) is an arbovirus maintained in the environment through a bird-mosquito enzootic cycle. Previous surveillance plans highlighted the endemicity of USUV in North-eastern Italy. In this work, we sequenced 138 new USUV full genomes from mosquito pools (Culex pipiens) and wild birds collected in North-eastern Italy and we investigated the evolutionary processes (phylogenetic analysis, selection pressure and evolutionary time-scale analysis) and spatial spread of USUV strains circulating in the European context and in Italy, with a particular focus on North-eastern Italy. Our results confirmed the circulation of viruses belonging to four different lineages in Italy (EU1, EU2, EU3 and EU4), with the newly sequenced viruses from the North-eastern regions, Veneto and Friuli Venezia Giulia, belonging to the EU2 lineage and clustering into two different sub-lineages, EU2-A and EU2-B. Specific mutations characterize each European lineage and geographic location seem to have shaped their phylogenetic structure. By investigating the spatial spread in Europe, we were able to show that Italy acted mainly as donor of USUV to neighbouring countries. At a national level, we identified two geographical clusters mainly circulating in Northern and North-western Italy, spreading both northward and southward. Our analyses provide important information on the spatial and evolutionary dynamics of USUTU virus that can help to improve surveillance plans and control strategies for this virus of increasing concern for human health.
Collapse
Affiliation(s)
- B Zecchin
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - A Fusaro
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - A Milani
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - A Schivo
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - S Ravagnan
- National Reference Centre/OIE Collaborating Centre for Diseases at the Animal-Human Interface, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - S Ormelli
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - C Mavian
- Emerging Pathogens Institute, Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - A Michelutti
- National Reference Centre/OIE Collaborating Centre for Diseases at the Animal-Human Interface, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - F Toniolo
- National Reference Centre/OIE Collaborating Centre for Diseases at the Animal-Human Interface, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - L Barzon
- Department of Molecular Medicine, University of Padua, Padova, Italy
| | - I Monne
- Department of Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - G Capelli
- National Reference Centre/OIE Collaborating Centre for Diseases at the Animal-Human Interface, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| |
Collapse
|
18
|
Castro A, Carreño JM, Duehr J, Krammer F, Kane RS. Refocusing the Immune Response to Selected Epitopes on a Zika Virus Protein Antigen by Nanopatterning. Adv Healthc Mater 2021; 10:e2002140. [PMID: 33929789 DOI: 10.1002/adhm.202002140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/27/2021] [Indexed: 12/11/2022]
Abstract
Infections with Zika virus (ZIKV) are linked to the development of severe central nervous system disorders, but the need for a ZIKV vaccine remains unmet. Although the design of vaccines that elicit antibodies targeting domain III (DIII) of the ZIKV envelope (E) protein as an antigen is an attractive strategy, poorly neutralizing or cross-reactive antibodies that target the E protein may lead to antibody-dependent enhancement of disease. It is therefore decided to use the previously reported nanopatterning technique, which combines the site-specific incorporation of non-canonical amino acids with site-specific functionalization of the protein with polyethylene glycol (PEG), to shield selected epitopes on DIII. Two different nanopatterned DIII variants are designed and characterized and demonstrate that epitope shielding with PEG completely inhibits the binding of epitope-specific antibodies in vitro. Furthermore, immunization with multivalent nanopatterned DIII antigens results in the refocusing of the antibody response toward the exposed epitopes on the protein surface and away from potentially enhancing epitopes. This ability to redirect the antibody response toward targeted regions of the DIII protein should be useful for the design of effective and safe ZIKV vaccines.
Collapse
Affiliation(s)
- Ana Castro
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology Atlanta GA 30332 USA
| | - Juan Manuel Carreño
- Department of Microbiology Icahn School of Medicine at Mount Sinai New York NY 10029 USA
| | - James Duehr
- Department of Microbiology Icahn School of Medicine at Mount Sinai New York NY 10029 USA
| | - Florian Krammer
- Department of Microbiology Icahn School of Medicine at Mount Sinai New York NY 10029 USA
| | - Ravi S. Kane
- School of Chemical and Biomolecular Engineering Georgia Institute of Technology Atlanta GA 30332 USA
| |
Collapse
|
19
|
Computational and Rational Design of Single-Chain Antibody against Tick-Borne Encephalitis Virus for Modifying Its Specificity. Viruses 2021; 13:v13081494. [PMID: 34452359 PMCID: PMC8402911 DOI: 10.3390/v13081494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/09/2021] [Accepted: 06/23/2021] [Indexed: 12/27/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) causes 5−7 thousand cases of human meningitis and encephalitis annually. The neutralizing and protective antibody ch14D5 is a potential therapeutic agent. This antibody exhibits a high affinity for binding with the D3 domain of the glycoprotein E of the Far Eastern subtype of the virus, but a lower affinity for the D3 domains of the Siberian and European subtypes. In this study, a 2.2-fold increase in the affinity of single-chain antibody sc14D5 to D3 proteins of the Siberian and European subtypes of the virus was achieved using rational design and computational modeling. This improvement can be further enhanced in the case of the bivalent binding of the full-length chimeric antibody containing the identified mutation.
Collapse
|
20
|
Fibriansah G, Lim XN, Lok SM. Morphological Diversity and Dynamics of Dengue Virus Affecting Antigenicity. Viruses 2021; 13:v13081446. [PMID: 34452312 PMCID: PMC8402850 DOI: 10.3390/v13081446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 01/30/2023] Open
Abstract
The four serotypes of the mature dengue virus can display different morphologies, including the compact spherical, the bumpy spherical and the non-spherical clubshape morphologies. In addition, the maturation process of dengue virus is inefficient and therefore some partially immature dengue virus particles have been observed and they are infectious. All these viral particles have different antigenicity profiles and thus may affect the type of the elicited antibodies during an immune response. Understanding the molecular determinants and environmental conditions (e.g., temperature) in inducing morphological changes in the virus and how potent antibodies interact with these particles is important for designing effective therapeutics or vaccines. Several techniques, including cryoEM, site-directed mutagenesis, hydrogen-deuterium exchange mass spectrometry, time-resolve fluorescence resonance energy transfer, and molecular dynamic simulation, have been performed to investigate the structural changes. This review describes all known morphological variants of DENV discovered thus far, their surface protein dynamics and the key residues or interactions that play important roles in the structural changes.
Collapse
Affiliation(s)
- Guntur Fibriansah
- Programme in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.F.); (X.-N.L.)
- Centre for BioImaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Xin-Ni Lim
- Programme in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.F.); (X.-N.L.)
- Centre for BioImaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore 117557, Singapore
| | - Shee-Mei Lok
- Programme in Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore 169857, Singapore; (G.F.); (X.-N.L.)
- Centre for BioImaging Sciences, Department of Biological Sciences, National University of Singapore, Singapore 117557, Singapore
- Correspondence:
| |
Collapse
|
21
|
Maciejewski S, Ruckwardt TJ, Morabito KM, Foreman BM, Burgomaster KE, Gordon DN, Pelc RS, DeMaso CR, Ko SY, Fisher BE, Yang ES, Nair D, Foulds KE, Todd JP, Kong WP, Roy V, Aleshnick M, Speer SD, Bourne N, Barrett AD, Nason MC, Roederer M, Gaudinski MR, Chen GL, Dowd KA, Ledgerwood JE, Alter G, Mascola JR, Graham BS, Pierson TC. Distinct neutralizing antibody correlates of protection among related Zika virus vaccines identify a role for antibody quality. Sci Transl Med 2021; 12:12/547/eaaw9066. [PMID: 32522807 DOI: 10.1126/scitranslmed.aaw9066] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 11/29/2019] [Accepted: 05/19/2020] [Indexed: 12/25/2022]
Abstract
The emergence of Zika virus (ZIKV) in the Americas stimulated the development of multiple ZIKV vaccine candidates. We previously developed two related DNA vaccine candidates encoding ZIKV structural proteins that were immunogenic in animal models and humans. We sought to identify neutralizing antibody (NAb) properties induced by each vaccine that correlated with protection in nonhuman primates (NHPs). Despite eliciting equivalent NAb titers in NHPs, these vaccines were not equally protective. The transfer of equivalent titers of vaccine-elicited NAb into AG129 mice also revealed nonequivalent protection, indicating qualitative differences among antibodies (Abs) elicited by these vaccines. Both vaccines elicited Abs with similar binding titers against envelope protein monomers and those incorporated into virus-like particles, as well as a comparable capacity to orchestrate phagocytosis. Functional analysis of vaccine-elicited NAbs from NHPs and humans revealed a capacity to neutralize the structurally mature form of the ZIKV virion that varied in magnitude among vaccine candidates. Conversely, sensitivity to the virion maturation state was not a characteristic of NAbs induced by natural or experimental infection. Passive transfer experiments in mice revealed that neutralization of mature ZIKV virions more accurately predicts protection from ZIKV infection. These findings demonstrate that NAb correlates of protection may differ among vaccine antigens when assayed using standard neutralization platforms and suggest that measurements of Ab quality, including the capacity to neutralize mature virions, will be critical for defining correlates of ZIKV vaccine-induced immunity.
Collapse
Affiliation(s)
| | | | | | - Bryant M Foreman
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - David N Gordon
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Rebecca S Pelc
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Sung-Youl Ko
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Brian E Fisher
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Eun Sung Yang
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Deepika Nair
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - John Paul Todd
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Wing-Pui Kong
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Vicky Roy
- Ragon Institute, Cambridge, MA 02139, USA
| | - Maya Aleshnick
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Scott D Speer
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | - Nigel Bourne
- Department of Microbiology and Immunology, Department of Pathology, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alan D Barrett
- Department of Microbiology and Immunology, Department of Pathology, Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Martha C Nason
- Biostatistics Research Branch, Division of Clinical Research, NIAID, NIH, Bethesda, MD 20852, USA
| | - Mario Roederer
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | | | - Grace L Chen
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Kimberly A Dowd
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20892, USA
| | | | | | - John R Mascola
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA
| | - Barney S Graham
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA.
| | | |
Collapse
|
22
|
Kotaki T, Kurosu T, Grinyo-Escuer A, Davidson E, Churrotin S, Okabayashi T, Puiprom O, Mulyatno KC, Sucipto TH, Doranz BJ, Ono KI, Soegijanto S, Kameoka M. An affinity-matured human monoclonal antibody targeting fusion loop epitope of dengue virus with in vivo therapeutic potency. Sci Rep 2021; 11:12987. [PMID: 34155267 PMCID: PMC8217507 DOI: 10.1038/s41598-021-92403-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/08/2021] [Indexed: 11/26/2022] Open
Abstract
Dengue virus (DENV), from the genus flavivirus of the family flaviviridae, causes serious health problems globally. Human monoclonal antibodies (HuMAb) can be used to elucidate the mechanisms of neutralization and antibody-dependent enhancement (ADE) of DENV infections, leading to the development of a vaccine or therapeutic antibodies. Here, we generated eight HuMAb clones from an Indonesian patient infected with DENV. These HuMAbs exhibited the typical characteristics of weak neutralizing antibodies including high cross-reactivity with other flaviviruses and targeting of the fusion loop epitope (FLE). However, one of the HuMAbs, 3G9, exhibited strong neutralization (NT50 < 0.1 μg/ml) and possessed a high somatic hyper-mutation rate of the variable region, indicating affinity-maturation. Administration of this antibody significantly prolonged the survival of interferon-α/β/γ receptor knockout C57BL/6 mice after a lethal DENV challenge. Additionally, Fc-modified 3G9 that had lost their in vitro ADE activity showed enhanced therapeutic potency in vivo and competed strongly with an ADE-prone antibody in vitro. Taken together, the affinity-matured FLE-targeting antibody 3G9 exhibits promising features for therapeutic application including a low NT50 value, potential for treatment of various kinds of mosquito-borne flavivirus infection, and suppression of ADE. This study demonstrates the therapeutic potency of affinity-matured FLE-targeting antibodies.
Collapse
Affiliation(s)
- Tomohiro Kotaki
- Department of Public Health, Kobe University Graduate School of Health Sciences, Kobe, Japan.
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia.
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan.
| | - Takeshi Kurosu
- Department of Virology I, National Institute of Infectious Diseases (NIID), Tokyo, Japan
| | | | | | - Siti Churrotin
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Tamaki Okabayashi
- Mahidol-Osaka Center for Infectious Diseases (MOCID), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Orapim Puiprom
- Mahidol-Osaka Center for Infectious Diseases (MOCID), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kris Cahyo Mulyatno
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Teguh Hari Sucipto
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | | | - Ken-Ichiro Ono
- Medical & Biological Laboratories Co., Ltd., Tokyo, Japan
| | - Soegeng Soegijanto
- Collaborative Research Center for Emerging and Re-Emerging Infectious Diseases, Institute of Tropical Disease, Airlangga University, Surabaya, Indonesia
| | - Masanori Kameoka
- Department of Public Health, Kobe University Graduate School of Health Sciences, Kobe, Japan.
| |
Collapse
|
23
|
Agudelo M, Palus M, Keeffe JR, Bianchini F, Svoboda P, Salát J, Peace A, Gazumyan A, Cipolla M, Kapoor T, Guidetti F, Yao KH, Elsterová J, Teislerová D, Chrdle A, Hönig V, Oliveira T, West AP, Lee YE, Rice CM, MacDonald MR, Bjorkman PJ, Růžek D, Robbiani DF, Nussenzweig MC. Broad and potent neutralizing human antibodies to tick-borne flaviviruses protect mice from disease. J Exp Med 2021; 218:e20210236. [PMID: 33831141 PMCID: PMC8040517 DOI: 10.1084/jem.20210236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/30/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV) is an emerging human pathogen that causes potentially fatal disease with no specific treatment. Mouse monoclonal antibodies are protective against TBEV, but little is known about the human antibody response to infection. Here, we report on the human neutralizing antibody response to TBEV in a cohort of infected and vaccinated individuals. Expanded clones of memory B cells expressed closely related anti-envelope domain III (EDIII) antibodies in both groups of volunteers. However, the most potent neutralizing antibodies, with IC50s below 1 ng/ml, were found only in individuals who recovered from natural infection. These antibodies also neutralized other tick-borne flaviviruses, including Langat, louping ill, Omsk hemorrhagic fever, Kyasanur forest disease, and Powassan viruses. Structural analysis revealed a conserved epitope near the lateral ridge of EDIII adjoining the EDI-EDIII hinge region. Prophylactic or early therapeutic antibody administration was effective at low doses in mice that were lethally infected with TBEV.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Neutralizing/administration & dosage
- Antibodies, Neutralizing/genetics
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/administration & dosage
- Antibodies, Viral/genetics
- Antibodies, Viral/immunology
- Cells, Cultured
- Cohort Studies
- Cross Reactions/immunology
- Encephalitis Viruses, Tick-Borne/drug effects
- Encephalitis Viruses, Tick-Borne/immunology
- Encephalitis Viruses, Tick-Borne/physiology
- Encephalitis, Tick-Borne/immunology
- Encephalitis, Tick-Borne/prevention & control
- Encephalitis, Tick-Borne/virology
- Epitopes/immunology
- Female
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/immunology
- Mice, Inbred BALB C
- Sequence Homology, Amino Acid
- Survival Analysis
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Mice
Collapse
Affiliation(s)
- Marianna Agudelo
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Martin Palus
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
- Veterinary Research Institute, Brno, Czech Republic
| | - Jennifer R. Keeffe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Filippo Bianchini
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Pavel Svoboda
- Veterinary Research Institute, Brno, Czech Republic
- Department of Pharmacology and Pharmacy, Faculty of Veterinary Medicine, University of Veterinary and Pharmaceutical Sciences Brno, Brno, Czech Republic
| | - Jiří Salát
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
- Veterinary Research Institute, Brno, Czech Republic
| | - Avery Peace
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Melissa Cipolla
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Tania Kapoor
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Francesca Guidetti
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Kai-Hui Yao
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Jana Elsterová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
- Veterinary Research Institute, Brno, Czech Republic
| | | | - Aleš Chrdle
- Hospital České Budějovice, České Budějovice, Czech Republic
- Faculty of Social and Health Sciences, University of South Bohemia, České Budějovice, Czech Republic
- Royal Liverpool University Hospital, Liverpool, UK
| | - Václav Hönig
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
- Veterinary Research Institute, Brno, Czech Republic
| | - Thiago Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
| | - Anthony P. West
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Yu E. Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Margaret R. MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Pamela J. Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Daniel Růžek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czech Republic
- Veterinary Research Institute, Brno, Czech Republic
| | - Davide F. Robbiani
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Michel C. Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY
| |
Collapse
|
24
|
Vang L, Morello CS, Mendy J, Thompson D, Manayani D, Guenther B, Julander J, Sanford D, Jain A, Patel A, Shabram P, Smith J, Alexander J. Zika virus-like particle vaccine protects AG129 mice and rhesus macaques against Zika virus. PLoS Negl Trop Dis 2021; 15:e0009195. [PMID: 33711018 PMCID: PMC7990201 DOI: 10.1371/journal.pntd.0009195] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/24/2021] [Accepted: 02/02/2021] [Indexed: 11/30/2022] Open
Abstract
Background Zika virus (ZIKV), a mosquito-borne flavivirus, is a re-emerging virus that constitutes a public health threat due to its recent global spread, recurrent outbreaks, and infections that are associated with neurological abnormalities in developing fetuses and Guillain-Barré syndrome in adults. To date, there are no approved vaccines against ZIKV infection. Various preclinical and clinical development programs are currently ongoing in an effort to bring forward a vaccine for ZIKV. Methodology/Principle findings We have developed a ZIKV vaccine candidate based on Virus-Like-Particles (VLPs) produced in HEK293 mammalian cells using the prM (a precursor to M protein) and envelope (E) structural protein genes from ZIKV. Transient transfection of cells via plasmid and electroporation produced VLPs which were subsequently purified by column chromatography yielding approximately 2mg/L. Initially, immunogenicity and efficacy were evaluated in AG129 mice using a dose titration of VLP with and without Alhydrogel 2% (alum) adjuvant. We found that VLP with and without alum elicited ZIKV-specific serum neutralizing antibodies (nAbs) and that titers correlated with protection. A follow-up immunogenicity and efficacy study in rhesus macaques was performed using VLP formulated with alum. Multiple neutralization assay methods were performed on immune sera including a plaque reduction neutralization test, a microneutralization assay, and a Zika virus Renilla luciferase neutralization assay. All of these assays indicate that following immunization, VLP induces high titer nAbs which correlate with protection against ZIKV challenge. Conclusions/Significance These studies confirm that ZIKV VLPs could be efficiently generated and purified. Upon VLP immunization, in both mice and NHPs, nAb was induced that correlate with protection against ZIKV challenge. These studies support translational efforts in developing a ZIKV VLP vaccine for evaluation in human clinical trials. Zika virus (ZIKV) is a significant global health threat particularly due to the speed in which epidemics can occur. The resulting infections have been demonstrated to harm a developing fetus and, in some adults, be a co-factor for the development of Guillain-Barré syndrome. ZIKV is typically spread by the Aedes mosquito, but sexual transmission is also possible. We sought to develop a ZIKV prophylactic vaccine based on surface glycoproteins of the virus that would be devoid of any viral genetic material. This Virus-Like-Particle (VLP) was generated in vitro following introduction of plasmid DNA encoding Zika structural protein (prM-E) genes into mammalian cells. The aluminum-adjuvanted VLP induced nAbs in mice and nonhuman primates and protected against ZIKV challenge in vivo. These studies support the evaluation of this VLP candidate vaccine in human clinical trials.
Collapse
Affiliation(s)
- Lo Vang
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
- * E-mail:
| | | | - Jason Mendy
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Danielle Thompson
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Darly Manayani
- PaxVax Inc., San Diego, California, United States of America (PaxVax was acquired by Emergent BioSolutions Inc. Oct 2018)
| | - Ben Guenther
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Justin Julander
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, Utah, United States of America
| | - Daniel Sanford
- Battelle Biomedical Research Center, West Jefferson, Ohio, United States of America
| | - Amit Jain
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Amish Patel
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Paul Shabram
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
| | - Jonathan Smith
- PaxVax Inc., San Diego, California, United States of America (PaxVax was acquired by Emergent BioSolutions Inc. Oct 2018)
| | - Jeff Alexander
- Emergent BioSolutions Inc., Gaithersburg, Maryland, United States of America
- PaxVax Inc., San Diego, California, United States of America (PaxVax was acquired by Emergent BioSolutions Inc. Oct 2018)
| |
Collapse
|
25
|
Substantial Attenuation of Virulence of Tembusu Virus Strain PS Is Determined by an Arginine at Residue 304 of the Envelope Protein. J Virol 2021; 95:JVI.02331-20. [PMID: 33328312 DOI: 10.1128/jvi.02331-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
The Tembusu virus (TMUV) PS strain, derived by several passages and plaque purifications in BHK-21 cells, displays markedly lower virulence in Pekin ducklings relative to a natural isolate of TMUV, but the potential virulence determinants and the in vivo mechanisms for substantial virulence attenuation of the passage variant remain unknown. Here, we constructed a series of chimeric and mutant viruses and assessed their virulence using a 2-day-old Pekin duckling model. We showed that residue 304 in the envelope (E) protein is the molecular determinant of TMUV virulence. Further investigations with mutant and parental viruses demonstrated that acquisition of positive charges at E protein residue 304 plays a critical role in substantial attenuation of neurovirulence and neuroinvasiveness, which is linked to enhanced binding affinity for glycosaminoglycans (GAGs). In Pekin ducklings infected by subcutaneous inoculation, an Arg at residue 304 in the E protein was shown to contribute to more rapid virus clearance from the circulation, markedly reduced viremia, and significantly decreased viral growth in the extraneural tissues and the central nervous system, relative to a Met at the corresponding residue. These findings suggest that the in vivo mechanism of virulence attenuation of the TMUV passage variant closely resembles that proposed previously for GAG-binding variants of other flaviviruses. Overall, our study provides insight into the molecular basis of TMUV virulence and the in vivo consequences of acquisition of a GAG-binding determinant at residue 304 in the E protein of TMUV.IMPORTANCE TMUV-related disease emerged in 2010 and has a significant economic impact on the duck industry. Although the disease was originally recognized to affect adult ducks, increasing evidence has shown that TMUV also causes severe disease of young ducklings. It is, therefore, essential to investigate the pathogenesis of TMUV infection in a young duckling model. The significance of our studies is in identifying E protein residue Arg304 as the molecular determinant for TMUV virulence and in clarifying the crucial role of positive charges at E protein residue 304 in virulence attenuation of a TMUV passage variant. These data will greatly enhance our understanding of the pathogenesis of TMUV infection in ducklings and have implications for development of a safe and efficient vaccine.
Collapse
|
26
|
Fibriansah G, Lim EXY, Marzinek JK, Ng TS, Tan JL, Huber RG, Lim XN, Chew VSY, Kostyuchenko VA, Shi J, Anand GS, Bond PJ, Crowe JE, Lok SM. Antibody affinity versus dengue morphology influences neutralization. PLoS Pathog 2021; 17:e1009331. [PMID: 33621239 PMCID: PMC7935256 DOI: 10.1371/journal.ppat.1009331] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 03/05/2021] [Accepted: 01/25/2021] [Indexed: 12/23/2022] Open
Abstract
Different strains within a dengue serotype (DENV1-4) can have smooth, or “bumpy” surface morphologies with different antigenic characteristics at average body temperature (37°C). We determined the neutralizing properties of a serotype cross-reactive human monoclonal antibody (HMAb) 1C19 for strains with differing morphologies within the DENV1 and DENV2 serotypes. We mapped the 1C19 epitope to E protein domain II by hydrogen deuterium exchange mass spectrometry, cryoEM and molecular dynamics simulations, revealing that this epitope is likely partially hidden on the virus surface. We showed the antibody has high affinity for binding to recombinant DENV1 E proteins compared to those of DENV2, consistent with its strong neutralizing activities for all DENV1 strains tested regardless of their morphologies. This finding suggests that the antibody could out-compete E-to-E interaction for binding to its epitope. In contrast, for DENV2, HMAb 1C19 can only neutralize when the epitope becomes exposed on the bumpy-surfaced particle. Although HMAb 1C19 is not a suitable therapeutic candidate, this study with HMAb 1C19 shows the importance of choosing a high-affinity antibody that could neutralize diverse dengue virus morphologies for therapeutic purposes. Dengue virus consists of four serotypes (DENV1-4) and there are different strains within a serotype. DENV can have smooth or bumpy surface morphologies at physiological body temperature of 37°C, depending on the strain. We have determined the cryoEM structures of a cross-reactive neutralizing human monoclonal antibody (HMAb) 1C19 in complex with strains of DENV1 and DENV2 that form either smooth or bumpy surface morphologies. We have mapped the epitope of HMAb 1C19 to E protein domain II and the epitope is likely partially hidden on the virus surface. We showed that the antibody has high affinity for binding to recombinant DENV1 E protein than to DENV2 E protein. This explains the strong neutralization activity for all DENV1 strains tested regardless of their morphologies at physiological temperature, whereas it can only neutralize DENV2 strain that exposes the epitope on the bumpy surface particles. These results suggest that high-affinity therapeutic antibodies could neutralize diverse dengue virus morphologies.
Collapse
Affiliation(s)
- Guntur Fibriansah
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Elisa X. Y. Lim
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Jan K. Marzinek
- Bioinformatics Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Thiam-Seng Ng
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Joanne L. Tan
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Roland G. Huber
- Bioinformatics Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Xin-Ni Lim
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Valerie S. Y. Chew
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Victor A. Kostyuchenko
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Jian Shi
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
| | - Ganesh S. Anand
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Peter J. Bond
- Bioinformatics Institute, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - James E. Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Departments of Pediatrics and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- * E-mail: (JEC); (SML)
| | - Shee-Mei Lok
- Emerging Infectious Diseases, Duke–National University of Singapore Medical School, Singapore, Singapore
- Centre for BioImaging Sciences, National University of Singapore, Singapore, Singapore
- * E-mail: (JEC); (SML)
| |
Collapse
|
27
|
Xue L, Ren X, Magpantay F, Sun W, Zhu H. Optimal Control of Mitigation Strategies for Dengue Virus Transmission. Bull Math Biol 2021; 83:8. [PMID: 33404917 DOI: 10.1007/s11538-020-00839-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 11/21/2020] [Indexed: 11/30/2022]
Abstract
Dengue virus is transmitted by Aedes mosquitoes, posing threat to people's health and leading to great economic cost in many tropical and subtropical regions. We develop an ordinary differential equation model taking into account multiple strains of dengue virus. Using the model, we assess the effectiveness of human vaccination considering its waning and failure. We derive the lower bound and upper bound for the final size of the epidemic. Sensitivity analysis quantifies the impact of parameters on the basic reproduction number. Different scenarios of vaccinating humans show that it is better to vaccinate humans at early stages. We find that the cumulative number of infected humans is small when the vaccination rate is high or the waning rate is low for previously infected humans. We analyze the necessary conditions for implementing optimal control and derive the corresponding optimal solutions for mitigation dengue virus transmission by applying Pontryagin's Maximum Principle. Our findings may provide guidance for the public health authorities to implement human vaccination and other mitigation strategies.
Collapse
Affiliation(s)
- Ling Xue
- College of Automation, Harbin Engineering University, Harbin, 150001, China.,College of Mathematical Sciences, Harbin Engineering University, Harbin, 150001, China
| | - Xue Ren
- College of Automation, Harbin Engineering University, Harbin, 150001, China.,College of Mathematical Sciences, Harbin Engineering University, Harbin, 150001, China
| | - Felicia Magpantay
- Department of Mathematics and Statistics, Queen's University, Kingston, K7L 3N6, Canada
| | - Wei Sun
- College of Mathematical Sciences, Harbin Engineering University, Harbin, 150001, China.
| | - Huaiping Zhu
- Department of Mathematics and Statistics, York University, Toronto, M3J 1P3, Canada
| |
Collapse
|
28
|
Assessing the risk of dengue severity using demographic information and laboratory test results with machine learning. PLoS Negl Trop Dis 2020; 14:e0008960. [PMID: 33362244 PMCID: PMC7757819 DOI: 10.1371/journal.pntd.0008960] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Dengue virus causes a wide spectrum of disease, which ranges from subclinical disease to severe dengue shock syndrome. However, estimating the risk of severe outcomes using clinical presentation or laboratory test results for rapid patient triage remains a challenge. Here, we aimed to develop prognostic models for severe dengue using machine learning, according to demographic information and clinical laboratory data of patients with dengue. METHODOLOGY/PRINCIPAL FINDINGS Out of 1,581 patients in the National Cheng Kung University Hospital with suspected dengue infections and subjected to NS1 antigen, IgM and IgG, and qRT-PCR tests, 798 patients including 138 severe cases were enrolled in the study. The primary target outcome was severe dengue. Machine learning models were trained and tested using the patient dataset that included demographic information and qualitative laboratory test results collected on day 1 when they sought medical advice. To develop prognostic models, we applied various machine learning methods, including logistic regression, random forest, gradient boosting machine, support vector classifier, and artificial neural network, and compared the performance of the methods. The artificial neural network showed the highest average discrimination area under the receiver operating characteristic curve (0.8324 ± 0.0268) and balance accuracy (0.7523 ± 0.0273). According to the model explainer that analyzed the contributions/co-contributions of the different factors, patient age and dengue NS1 antigenemia were the two most important risk factors associated with severe dengue. Additionally, co-existence of anti-dengue IgM and IgG in patients with dengue increased the probability of severe dengue. CONCLUSIONS/SIGNIFICANCE We developed prognostic models for the prediction of dengue severity in patients, using machine learning. The discriminative ability of the artificial neural network exhibited good performance for severe dengue prognosis. This model could help clinicians obtain a rapid prognosis during dengue outbreaks. However, the model requires further validation using external cohorts in future studies.
Collapse
|
29
|
Lv J, Liu X, Cui S, Yang L, Qu S, Meng R, Yang B, Feng C, Wang X, Zhang D. The Neutralizing Antibody Response Elicited by Tembusu Virus Is Affected Dramatically by a Single Mutation in the Stem Region of the Envelope Protein. Front Microbiol 2020; 11:585194. [PMID: 33193231 PMCID: PMC7642334 DOI: 10.3389/fmicb.2020.585194] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/02/2020] [Indexed: 12/30/2022] Open
Abstract
Tembusu virus (TMUV) is a mosquito-borne flavivirus that most commonly affects adult breeder and layer ducks. However, a TMUV-caused neurological disease has also been found in ducklings below 7 weeks of age, highlighting the need to develop a safe vaccine for young ducklings. In this study, a plaque-purified PS TMUV strain was attenuated by serial passage in BHK-21 cells. Using 1-day-old Pekin ducklings as a model, the virus was confirmed to be attenuated sufficiently after 180 passages, whereas the neutralizing antibody response elicited by the 180th passage virus (PS180) was substantially impaired compared with PS. The findings suggest that sufficient attenuation results in loss of immunogenicity in the development of the live-attenuated TMUV vaccine. Comparative sequence analysis revealed that PS180 acquired one mutation (V41M) in prM and four mutations (T70A, Y176H, K313R, and F408L) in the envelope (E) protein. To identify the amino acid substitution(s) associated with loss of immunogenicity of PS180, we rescued parental viruses, rPS and rPS180, and produced mutant viruses, rPS180-M41V, rPS180-A70T, rPS180-H176Y, rPS180-R313K, rPS180-L408F, and rPS180-M5, which contained residue 41V in prM, residues 70T, 176Y, 313K, and 408F in E, and combination of the five residues, respectively, of PS in the backbone of the rPS180 genome. The neutralizing antibody response elicited by rPS180-L408F and rPS180-M5 was significantly higher than those by other mutant viruses and comparable to that by rPS. Furthermore, we produced mutant virus rPS-F408L, which contained residue 408L of PS180 in the backbone of the rPS genome. The F408L mutation conferred significantly decreased neutralizing antibody response to rPS-F408L, which was comparable to that elicited by rPS180. Based on homologous modeling, residue 408 was predicted to be located within the first helical domain of the stem region of the E protein (EH1). Together, these data demonstrate that a single mutation within the EH1 domain exerts a dramatical impact on the TMUV neutralizing antibody response. The present work may enhance our understanding of molecular basis of the TMUV neutralizing antibody response, and provides an important step for the development of a safe and efficient live-attenuated TMUV vaccine.
Collapse
Affiliation(s)
- Junfeng Lv
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoxiao Liu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shulin Cui
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lixin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Shenghua Qu
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Runze Meng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Baolin Yang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Chonglun Feng
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoyan Wang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dabing Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
30
|
Kumar YN, Jeyakodi G, Kumar NP, Gunasekaran K, Jambulingam P. Molecular modelling analysis of T219A mutant envelope protein revealed novel virulence enhancing factors in Dengue virus isolated from Kerala state, India. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 195:105481. [PMID: 32497770 DOI: 10.1016/j.cmpb.2020.105481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/11/2020] [Accepted: 03/26/2020] [Indexed: 06/11/2023]
Abstract
Dengue virus (DENV) is an emerging health threat and its envelope glycoprotein E, is involved in the anchoring and fusion mechanisms. Anchoring followed by conformational changes of E-protein are responsible for the fusion and entry of DENV into host. The variation in the conformation of the E-protein due to mutations, results in its altered binding with antibodies (Abs) and also its receptors. This leads to failure of neutralization of DENV and enhance the infection. In our earlier studies we have identified T219A mutation in the E-protein of DENV and the present study is focused on the impact of this mutation on the conformation of E-protein and also its binding variation with Abs and Fc-γ receptor. A comparative molecular modelling studies of wild type and T219A mutant E-proteins revealed that, the mutation induced several conformational variations in the E-protein and resulted in the variable binding orientation with altered affinities. Further, the mutation was also observed to enhance the fusion mechanism by Fc-γ receptors that mediate the efficient entry of DENV into host cell through altered membrane fusion mechanism. Such conformational variations of E-protein could be the responsible factors for enhanced virulence of DENV infections.
Collapse
Affiliation(s)
- Y Nanda Kumar
- Biomedical Informatics Centre, Vector Control Research Center, Indian Council of Medical Research, Pondicherry, India, 605006; Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | - G Jeyakodi
- Biomedical Informatics Centre, Vector Control Research Center, Indian Council of Medical Research, Pondicherry, India, 605006
| | - N Pradeep Kumar
- Biomedical Informatics Centre, Vector Control Research Center, Indian Council of Medical Research, Pondicherry, India, 605006
| | - K Gunasekaran
- Biomedical Informatics Centre, Vector Control Research Center, Indian Council of Medical Research, Pondicherry, India, 605006
| | - P Jambulingam
- Biomedical Informatics Centre, Vector Control Research Center, Indian Council of Medical Research, Pondicherry, India, 605006
| |
Collapse
|
31
|
Katzelnick LC, Bos S, Harris E. Protective and enhancing interactions among dengue viruses 1-4 and Zika virus. Curr Opin Virol 2020; 43:59-70. [PMID: 32979816 DOI: 10.1016/j.coviro.2020.08.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 07/31/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022]
Abstract
Dengue viruses 1-4 (DENV 1-4) and Zika virus (ZIKV) are closely related flaviviruses transmitted by Aedes mosquitoes that co-circulate in Asia, the Americas, Africa, and Oceania. Here, we review recent and historical literature on in vitro experiments, animal models, and clinical and epidemiological studies to describe how the sequence of DENV 1-4 and ZIKV infections modulates subsequent dengue and Zika disease outcome. Overall, we find these interactions are asymmetric. Immunity from a prior DENV infection or a prior ZIKV infection can enhance future severe dengue disease for some DENV serotypes while protecting against other serotypes. Further, prior DENV immunity has not been shown to enhance future uncomplicated or severe Zika and instead appears to be protective. Interestingly, secondary ZIKV infection induces type-specific ZIKV immunity but only generates weakly cross-neutralizing anti-DENV/ZIKV immunity, consistent with risk of future dengue disease. In contrast, secondary DENV infection induces strongly cross-neutralizing antibodies that protect against subsequent severe dengue disease. These immunologic interactions may be explained by differences in virion structure between DENV 1-4 and ZIKV, which modulate thermostability, susceptibility to neutralization, and cell infectivity. Overall, these observations are important for the understanding and prediction of epidemics and the development and evaluation of dengue and Zika vaccines.
Collapse
Affiliation(s)
- Leah C Katzelnick
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| | - Sandra Bos
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, 185 Li Ka Shing Center, 1951 Oxford Street, Berkeley, CA 94720-3370, United States.
| |
Collapse
|
32
|
Kubinski M, Beicht J, Gerlach T, Volz A, Sutter G, Rimmelzwaan GF. Tick-Borne Encephalitis Virus: A Quest for Better Vaccines against a Virus on the Rise. Vaccines (Basel) 2020; 8:E451. [PMID: 32806696 PMCID: PMC7564546 DOI: 10.3390/vaccines8030451] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Tick-borne encephalitis virus (TBEV), a member of the family Flaviviridae, is one of the most important tick-transmitted viruses in Europe and Asia. Being a neurotropic virus, TBEV causes infection of the central nervous system, leading to various (permanent) neurological disorders summarized as tick-borne encephalitis (TBE). The incidence of TBE cases has increased due to the expansion of TBEV and its vectors. Since antiviral treatment is lacking, vaccination against TBEV is the most important protective measure. However, vaccination coverage is relatively low and immunogenicity of the currently available vaccines is limited, which may account for the vaccine failures that are observed. Understanding the TBEV-specific correlates of protection is of pivotal importance for developing novel and improved TBEV vaccines. For affording robust protection against infection and development of TBE, vaccines should induce both humoral and cellular immunity. In this review, the adaptive immunity induced upon TBEV infection and vaccination as well as novel approaches to produce improved TBEV vaccines are discussed.
Collapse
Affiliation(s)
- Mareike Kubinski
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Jana Beicht
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Thomas Gerlach
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany;
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-University (LMU) Munich, Veterinaerstr. 13, 80539 Munich, Germany;
| | - Guus F. Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Foundation (TiHo), Buenteweg 17, 30559 Hannover, Germany; (M.K.); (J.B.); (T.G.)
| |
Collapse
|
33
|
Sequential immunization induces strong and broad immunity against all four dengue virus serotypes. NPJ Vaccines 2020; 5:68. [PMID: 32728482 PMCID: PMC7382454 DOI: 10.1038/s41541-020-00216-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 11/24/2022] Open
Abstract
A major challenge in dengue vaccine development is the need to induce immunity against four dengue (DENV) serotypes. Dengvaxia®, the only licensed dengue vaccine, consists of four variant dengue antigens, one for each serotype. Three doses of immunization with the tetravalent vaccine induced only suboptimal protection against DENV1 and DENV2. Furthermore, vaccination paradoxically and adversely primes dengue naïve subjects to more severe dengue. Here, we have tested whether sequential immunization induces stronger and broader immunity against four DENV serotypes than tetravalent-formulated immunization. Mice were immunized with four DNA plasmids, each encoding the pre-membrane and envelope from one DENV serotype, either sequentially or simultaneously. The sequential immunization induced significantly higher levels of interferon (IFN)γ- or tumor necrosis factor (TNF)α-expressing CD4+ and CD8+ T cells to both serotype-specific and conserved epitopes than tetravalent immunization. Moreover, sequential immunization induced higher levels of neutralizing antibodies to all four DENV serotypes than tetravalent vaccination. Consistently, sequential immunization resulted in more diversified immunoglobulin repertoire, including increased complementarity determining region 3 (CDR3) length and more robust germinal center reactions. These results show that sequential immunization offers a simple approach to potentially overcome the current challenges encountered with tetravalent-formulated dengue vaccines.
Collapse
|
34
|
Matveev A, Matveev L, Stronin O, Baykov I, Emeljanova L, Khlusevich Y, Tikunova N. Characterization of neutralizing monoclonal antibody against tick-borne encephalitis virus in vivo. Vaccine 2020; 38:4309-4315. [PMID: 32409136 DOI: 10.1016/j.vaccine.2020.04.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/30/2022]
Abstract
Tick-borne encephalitis virus (TBEV) is the most important tick-transmitted pathogen in the family Flaviviridae and causes one of the most severe human neuroinfections. In this study, a neutralizing mouse mAb 14D5, which was previously shown to have cross-reactive binding to several flaviviruses belonging to the TBEV group, was examined for its prophylactic and therapeutic effects in BALB/c mice infected with TBEV. Before and after infection, mice were administrated mAb 14D5 at doses 100 μg and 10 μg per mouse. mAb 14D5 showed clear protective efficacy when injected at the high dose one day after infection, with survival rates that were TBEV dose-dependent. Prophylactic administration of mAb 14D5 was more effective than post-exposure administration and complete protection was documented when the mAb was administered one day before infection. The protective efficacy of mAb 14D5 was significantly higher than that of the anti-TBE serum immunoglobulin. However, no protection was observed in mice received the low dose of mAb 14D5 independent of the timing of mAb injection and TBEV dose. The ability of species-matched mAb 14D5 to mediate TBEV infection in mice was also investigated, and the results indicated that mAb 14D5 did not augment TBEV infection independent of the time of mAb administration. The neutralizing epitope for mAb 14D5 was localized in domain III of glycoprotein E of TBEV in a region between residues 301-339, which is conserved among flaviviruses from the TBEV group.
Collapse
Affiliation(s)
- Andrey Matveev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Leonid Matveev
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Oleg Stronin
- Russian Federal State Unitary Company "Microgen Scientific Industrial Company for Immunobiological Medicines" of the Health Ministry of Russian Federation, Branch in Tomsk 634040, Russia
| | - Ivan Baykov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Ljudmila Emeljanova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yana Khlusevich
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nina Tikunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia.
| |
Collapse
|
35
|
Hirsch J, Faber BW, Crowe JE, Verstrepen B, Cornelissen G. E. coli production process yields stable dengue 1 virus-sized particles (VSPs). Vaccine 2020; 38:3305-3312. [PMID: 32197924 DOI: 10.1016/j.vaccine.2020.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/26/2020] [Accepted: 03/02/2020] [Indexed: 12/25/2022]
Abstract
Dengue fever is one of the most wide-spread vector-borne diseases in the world. Although dengue-associated mortality is low, morbidity and economic impact are high. Current licensed vaccines are limited and mediate only partial protection, thus a cost-effective vaccine with improved efficacy is strongly needed. In this work, recombinant dengue serotype 1 E protein was produced in E. coli, inclusion bodies were isolated and the E protein solubilized in urea and purified using an immobilized metal chelate affinity column. The protein was refolded by dialysis in order to obtain virus-like particles (VLPs). Particle assembly was confirmed using size-exclusion chromatography, dynamic light scattering (DLS), transmission electron microscopy (TEM), atomic force microscopy and stimulated emission depletion fluorescence (STED) microscopy. Particle diameter was strongly dependent on temperature, pH, buffer salt composition, and addition of L-arginine. Particles were stable in carbonate buffer at pH 9.5 and higher at 4 °C and did not aggregate during short-term temperature increase up to 55 °C. However, on basis of the above analyses, especially the results of DLS, TEM and STED, it was concluded that the particles obtained did not have an optimal virus-like structure and were therefore designated "virus-sized particles" (VSPs) rather than VLPs. Immunization of rabbits with the particles did not induce neutralizing antibodies, despite the recognition of the native virus by rabbit antibodies. As the titers against the immunogen were much higher than against the (heat-inactivated) virus, it is assumed that the conformation of the particles at the time of immunization was not optimal. Studies are currently underway to improve the quality of the E protein virus-sized particles towards true virus-like particles in order to optimize its potential as a dengue vaccine candidate.
Collapse
Affiliation(s)
- Janet Hirsch
- Hamburg University of Applied Sciences, Ulmenliet 20, 21033 Hamburg, Germany.
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, the Netherlands.
| | - James E Crowe
- Departments of Pediatrics and Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 2213 Garland Avenue, Nashville, TN 37232-0417, USA.
| | - Babs Verstrepen
- Department of Virology, Biomedical Primate Research Centre, Lange Kleiweg 161, 2288 GJ Rijswijk, the Netherlands.
| | - Gesine Cornelissen
- Hamburg University of Applied Sciences, Ulmenliet 20, 21033 Hamburg, Germany.
| |
Collapse
|
36
|
Experimental Usutu Virus Infection in Domestic Canaries Serinus canaria. Viruses 2020; 12:v12020164. [PMID: 32023880 PMCID: PMC7077186 DOI: 10.3390/v12020164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 01/27/2020] [Indexed: 02/06/2023] Open
Abstract
Usutu virus (USUV) is a neurotropic flavivirus closely related to West Nile virus (WNV). Its enzootic cycle mainly involves mosquitoes and birds. Human infection can occur with occasional, but sometimes severe, neurological complications. Since its emergence and spread in Europe over the last two decades, USUV has been linked to significant avian outbreaks, especially among Passeriformes, including European blackbirds (Turdus merula). Strikingly, no in vivo avian model exists so far to study this arbovirus. The domestic canary (Serinus canaria) is a passerine, which is considered as a highly susceptible model of infection by WNV. Here, we experimentally challenged domestic canaries with two different doses of USUV. All inoculated birds presented detectable amounts of viral RNA in the blood and RNA shedding via feathers and droppings during the early stages of the infection, as determined by RT-qPCR. Mortality occurred in both infected groups (1/5 and 2/5, respectively) and was not necessarily correlated to a pure neurological disease. Subsequent analyses of samples from dead birds showed histopathological changes and virus tropism mimicking those reported in naturally infected birds. A robust seroconversion followed the infection in almost all the surviving canaries. Altogether, these results demonstrate that domestic canaries constitute an interesting experimental model for the study of USUV pathogenesis and transmission.
Collapse
|
37
|
Chaintoutis SC, Papa A, Pervanidou D, Dovas CI. Evolutionary dynamics of lineage 2 West Nile virus in Europe, 2004–2018: Phylogeny, selection pressure and phylogeography. Mol Phylogenet Evol 2019; 141:106617. [DOI: 10.1016/j.ympev.2019.106617] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/29/2022]
|
38
|
Versiani AF, Rocha RP, Mendes TAO, Pereira GC, Coelho dos Reis JGA, Bartholomeu DC, da Fonseca FG. Identification of B-Cell Epitopes with Potential to Serologicaly Discrimnate Dengue from Zika Infections. Viruses 2019; 11:E1079. [PMID: 31752352 PMCID: PMC6893796 DOI: 10.3390/v11111079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022] Open
Abstract
Dengue is currently one of the most important arbovirus infections worldwide. Early diagnosis is important for disease outcome, particularly for those afflicted with the severe forms of infection. The goal of this work was to identify conserved and polymorphic linear B-cell Dengue virus (DENV) epitopes that could be used for diagnostic purposes. To this end, we aligned the predicted viral proteome of the four DENV serotype and performed in silico B-cell epitope mapping. We developed a script in Perl integrating alignment and prediction information to identify potential serotype-specific epitopes. We excluded epitopes that were similarly present in the yellow fever and zika viruses' proteomes. A total of 15 polymorphic and nine conserved peptides among DENV serotypes were selected. Peptides were spotted on cellulose membranes and tested against sera from rabbits that were monoinfected with each DENV serotype. Although serotype-specific peptides failed to recognize any sera, three conserved peptides were recognized by all anti-dengue sera and were included on an ELISA test employing a well-characterized human sera bank. Of the three peptides, one was able to efficiently identify sera from all four DENV serotypes and to discriminate them from Zika virus positive sera.
Collapse
Affiliation(s)
- Alice F. Versiani
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (A.F.V.); (R.P.R.); (J.G.A.C.d.R.)
- Laboratório de Pesquisa em Virologia, Departamento de Doenças Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto 15090-000, Brazil
| | - Raissa Prado Rocha
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (A.F.V.); (R.P.R.); (J.G.A.C.d.R.)
| | - Tiago A. O. Mendes
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (T.A.O.M.); (D.C.B.)
| | | | - Jordana Graziella A. Coelho dos Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (A.F.V.); (R.P.R.); (J.G.A.C.d.R.)
| | - Daniella C. Bartholomeu
- Laboratório de Imunologia e Genômica de Parasitos, Departamento de Parasitologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (T.A.O.M.); (D.C.B.)
| | - Flávio G. da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (A.F.V.); (R.P.R.); (J.G.A.C.d.R.)
| |
Collapse
|
39
|
The truncated E protein of DTMUV provide protection in young ducks. Vet Microbiol 2019; 240:108508. [PMID: 31902493 DOI: 10.1016/j.vetmic.2019.108508] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/16/2019] [Accepted: 11/12/2019] [Indexed: 02/06/2023]
Abstract
Duck Tembusu virus (DTMUV) is a major pathogen of duck industry in China. In the current study, we generated different constructs containing envelope (E) protein, pre-membrane-envelope (prM-E) protein, and C-terminally truncated E protein of the DTMUV. The constructed proteins could induce specific antibody responses in young ducks. When ducklings were immunized with the constructed proteins, they were 100% protected against DTMUV infection. Furthermore, the fluorescent signal of the truncated E protein was stronger than other constructed proteins, when Bac-to-Bac baculovirus expression system was applied. Our data demonstrated that the truncated E protein used in the current study could be applied as a potential vaccine candidate to control DTMUV infection in young ducks.
Collapse
|
40
|
Davis EH, Barrett ADT. Structure-Function of the Yellow Fever Virus Envelope Protein: Analysis of Antibody Epitopes. Viral Immunol 2019; 33:12-21. [PMID: 31682201 DOI: 10.1089/vim.2019.0107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Yellow fever virus (YFV) is the prototype member of the genus Flavivirus, which contains more than 60 positive-sense, single-stranded RNA viruses, many of which are considered public health threats. YF disease is controlled by a live attenuated vaccine, 17D, which was generated empirically through serial passage of the wild-type (WT) strain Asibi in chicken tissue. The vaccine, which has been used for over 80 years, is considered to be one of the safest and most effective live attenuated vaccines. It has been shown that the humoral immune response is essential to a positive disease outcome during infection. As such, the neutralizing antibody response and its correlation to long-term protection are a critical measure of 17D efficacy. The primary target of these antibodies is the envelope (E) protein, which is the major component of the virion. Monoclonal antibodies can distinguish WT strain Asibi and vaccine strain 17D by many different measures, including physical binding, hemagglutination inhibition, neutralization, and passive protection. This makes the WT-vaccine pair ideal candidates to study the structure-function relationship of the E protein in the attenuation and immunogenicity of flaviviruses. In this study, we provide an overview of structure-function of YFV E protein and its involvement in protective immunity.
Collapse
Affiliation(s)
- Emily H Davis
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas.,Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
41
|
Galula JU, Salem GM, Chang GJJ, Chao DY. Does structurally-mature dengue virion matter in vaccine preparation in post-Dengvaxia era? Hum Vaccin Immunother 2019; 15:2328-2336. [PMID: 31314657 PMCID: PMC6816432 DOI: 10.1080/21645515.2019.1643676] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The unexpectedly low vaccine efficacy of Dengvaxia®, developed by Sanofi Pasteur, and a higher risk of severe diseases after vaccination among dengue-naive children or children younger than 6 years old, have cast skepticism about the safety of dengue vaccination resulting in the suspension of school-based immunization programs in the Philippines. The absence of immune correlates of protection from dengue virus (DENV) infection hampers the development of other potential DENV vaccines. While tetravalent live-attenuated tetravalent vaccines (LATVs), which mimic natural infection by inducing both cellular and humoral immune responses, are still currently favored, developing a vaccine that provides a balanced immunity to all four DENV serotypes remains a challenge. With the recently advanced understanding of virion structure and B cell immune responses from naturally infected DENV patients, two points of view in developing a next-generation dengue vaccine emerged: one is to induce potent, type-specific neutralizing antibodies (NtAbs) recognizing quaternary structure-dependent epitopes by having four components of vaccine strains replicate equivalently; the other is to induce protective and broadly NtAbs against the four serotypes of DENV with a universal vaccine. This article reviews the studies related to these issues and the current knowledge gap that needs to be filled in.
Collapse
Affiliation(s)
- Jedhan Ucat Galula
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Gielenny M Salem
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| | - Gwong-Jen J Chang
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, US Department of Health and Human Services , Fort Collins , CO , USA
| | - Day-Yu Chao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University , Taichung , Taiwan
| |
Collapse
|
42
|
Pinto PBA, Assis ML, Vallochi AL, Pacheco AR, Lima LM, Quaresma KRL, Pereira BAS, Costa SM, Alves AMB. T Cell Responses Induced by DNA Vaccines Based on the DENV2 E and NS1 Proteins in Mice: Importance in Protection and Immunodominant Epitope Identification. Front Immunol 2019; 10:1522. [PMID: 31333657 PMCID: PMC6617960 DOI: 10.3389/fimmu.2019.01522] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 06/18/2019] [Indexed: 01/04/2023] Open
Abstract
The importance of the cellular immune response against DENV has been increasingly highlighted in the past few years, in particular for vaccine development. We have previously constructed two plasmids, pE1D2, and pcTPANS1, encoding the envelope (E) ectodomain (domains I, II, and III) and the non-structural 1 (NS1) protein of dengue virus serotype 2 (DENV2), respectively. In the present work, we analyzed the induction of the cellular response in mice immunized with these DNA vaccines and identified the immunogenic peptides. Vaccinated BALB/c mice became protected against a lethal challenge of DENV2. Depletion of CD4+ cells in vaccinated animals almost completely abolished protection elicited by both vaccines. In contrast, a significant number of pE1D2- and pcTPANS1-immunized mice survived virus challenge after depletion of CD8+ cells, although some animals presented morbidity. To identify immunogenic peptides recognized by T cells, we stimulated splenocytes with overlapping peptide libraries covering the E and NS1 proteins and evaluated the production of IFN-γ by ELISPOT. We detected two and three immunodominant epitopes in the E and NS1 proteins, respectively, and four additional NS1-derived peptides after virus challenge. Characterization by intracellular cytokine staining (ICS) revealed that both CD4+ and CD8+ T cells were involved in IFN-γ and TNF-α production. The IFN-γ ICS confirmed reaction of almost all E-derived peptides before challenge and identified other epitopes after infection. All NS1-derived peptides were able to elicit IFN-γ production in CD4+ cells, while only a few peptides induced expression of this cytokine in CD8+ T lymphocytes. Interestingly, we observed an increase in the frequency of either CD4+ or CD8+ T cells producing TNF-α after immunization with the pE1D2 and challenge with DENV2, while lymphocytes from pcTPANS1-vaccinated animals maintained ordinary TNF-α production after virus infection. We also assessed the recognition of E and NS1 immunogenic peptides in C57BL/6 mice due to the difference in MHC haplotype expression. Two NS1-derived epitopes featured prominently in the IFN-γ response with cells from both animal strains. Overall, our results emphasize the importance of the T cell response involved in protection against dengue induced by E and NS1 based DNA vaccines.
Collapse
Affiliation(s)
- Paolla B. A. Pinto
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Maysa L. Assis
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Adriana L. Vallochi
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Agatha R. Pacheco
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Lauro M. Lima
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Kátia R. L. Quaresma
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Bernardo A. S. Pereira
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Simone M. Costa
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Ada M. B. Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Hou J, Shrivastava S, Fraser CC, Loo HL, Wong LH, Ho V, Fink K, Ooi EE, Chen J. Dengue Mosaic Vaccines Enhance Cellular Immunity and Expand the Breadth of Neutralizing Antibody Against All Four Serotypes of Dengue Viruses in Mice. Front Immunol 2019; 10:1429. [PMID: 31281322 PMCID: PMC6596366 DOI: 10.3389/fimmu.2019.01429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/06/2019] [Indexed: 11/13/2022] Open
Abstract
An estimated 400 million people in the world are infected with any of the four types of dengue virus (DENV) annually. The only licensed dengue vaccine cannot effectively prevent infection with all of the four DENVs, especially in those immunologically naïve at baseline. In this study, we explored a mosaic vaccine approach, which utilizes an artificial recombinant sequence for each serotype to achieve maximum coverage of variant epitopes in the four DENVs. We determined the immunogenicity and cross-reactivity of DNA plasmids encoding individual mosaic sequences or the natural sequences in mice. We show that the mosaic vaccines, particularly those targeting DENV serotype 1 and 2, improved vaccine immunogenicity by increasing the percentage of antigen-specific IFNγ- or TNFα-secreting CD4 and CD8 T cells, and titers of neutralizing antibodies. The mosaic vaccine diversified and broadened anti-dengue T cell responses and cross-reactive neutralizing antibodies against all four serotypes. The mosaic vaccines also induced higher level of antigen-specific B cell responses. These results suggest that mosaic vaccines comprising of DENV serotype 1 and 2 variant epitopes could stimulate strong and broad immune responses against all four serotypes.
Collapse
Affiliation(s)
- Jue Hou
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Shubham Shrivastava
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Christopher C Fraser
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Hooi Linn Loo
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Lan Hiong Wong
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Victor Ho
- Singapore Immunology Network, AStar, Singapore, Singapore
| | - Katja Fink
- Singapore Immunology Network, AStar, Singapore, Singapore
| | - Eng Eong Ooi
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.,Emerging Infectious Diseases Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore.,Koch Institute for Integrative Cancer Research and Departments of Biology, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
44
|
Sasmal SK, Takeuchi Y, Nakaoka S. T-Cell mediated adaptive immunity and antibody-dependent enhancement in secondary dengue infection. J Theor Biol 2019; 470:50-63. [DOI: 10.1016/j.jtbi.2019.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/13/2019] [Accepted: 03/12/2019] [Indexed: 11/26/2022]
|
45
|
Yamanaka A, Konishi E. Key Amino Acid Substitution for Infection-Enhancing Activity-Free Designer Dengue Vaccines. iScience 2019; 13:125-137. [PMID: 30826727 PMCID: PMC6402262 DOI: 10.1016/j.isci.2019.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 11/17/2022] Open
Abstract
Dengue is a globally important disease caused by four serotypes of dengue virus. Dengue vaccine development has been hampered by antigenic cross-reactivity among serotypes, which potentially causes antibody-dependent enhancement of infection and disease severity. Here we found that a single amino acid substitution in the envelope protein at position 87 from aspartic acid to asparagine or at position 107 from leucine to phenylalanine is critical for suppressing the induction of infection-enhancing antibody in a mouse model. The site and type of amino acid substitution were determined via neutralization escape using an enhancing-activity-only monoclonal antibody that was engineered to reveal neutralizing activity. Mutated dengue type 1 DNA vaccines containing either or both amino acid substitutions induced neutralizing antibodies devoid of enhancing activity against all serotypes. The effect of substitution was further demonstrated using other serotypes and a tetravalent formulation. This finding may contribute to the development of infection-enhancing-activity-free dengue vaccines. Amino acids at E87 or E107 are critical for dengue-enhancing antibody induction Neutralization escape is useful for identifying the key types or sites of amino acids Each substitution can be applied to antigens of all four dengue serotypes A modified tetravalent DNA vaccine suppresses enhancing antibody induction in mice
Collapse
Affiliation(s)
- Atsushi Yamanaka
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan.
| | - Eiji Konishi
- BIKEN Endowed Department of Dengue Vaccine Development, Faculty of Tropical Medicine, Mahidol University, 420/6 Ratchawithi Road, Ratchathewi, Bangkok 10400, Thailand; BIKEN Endowed Department of Dengue Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
46
|
Ul-Rahman A. Genetic diversity of Alkhurma hemorrhagic fever virus in Western Asia. INFECTION GENETICS AND EVOLUTION 2019; 70:80-83. [PMID: 30779959 DOI: 10.1016/j.meegid.2019.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 01/31/2019] [Accepted: 02/15/2019] [Indexed: 11/19/2022]
Abstract
Alkhurma hemorrhagic fever, caused by Alkhurma hemorrhagic fever virus (ALKV), is an arboviral infection which is further expanding in tropical and subtropical regions of the Western Asia. A number of Alkhurma hemorrhagic fever virus (ALKV) strains have been isolated from clinical cases representing Saudi Arabia and Egypt; however, the phylogenetic relationship of these particular isolates to those reported previously elsewhere in the world remains elusive. Based on the analysis of the envelope (E), and non-structural gene (NS3 and NS5), the phylogenetic and PASC analysis revealed the circulation of three sub-lineages (I-III) suggesting a continuous evolution. Also, the comparative genome analysis revealed the envelope gene to be a reliable genetic marker to elucidate the molecular epidemiology and genetic diversity of discrete strains of ALKV.
Collapse
Affiliation(s)
- Aziz Ul-Rahman
- Department of Microbiology and Quality Control Laboratory, University of Veterinary and Animal Lahore, 54000, Pakistan.
| |
Collapse
|
47
|
The distribution of important sero-complexes of flaviviruses in Malaysia. Trop Anim Health Prod 2019; 51:495-506. [PMID: 30604332 DOI: 10.1007/s11250-018-01786-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 12/13/2018] [Indexed: 12/13/2022]
Abstract
Flaviviruses (FVs) are arthropod-borne viruses of medical and veterinary importance. Numerous species of FVs have been isolated from various host; mainly humans, animals, ticks, and mosquitoes. Certain FVs are extremely host-specific; at the same time, some FVs can infect an extensive range of species. Based on published literatures, 11 species of FVs have been detected from diverse host species in Malaysia. In humans, dengue virus and Japanese encephalitis virus have been reported since 1901 and 1942. In animals, the Batu Cave virus, Sitiawan virus, Carey Island, Tembusu virus, Duck Tembusu virus, and Japanese encephalitis viruses were isolated from various species. In mosquitoes, Japanese encephalitis virus and Kunjin virus were isolated from Culex spp., while Zika virus and Jugra virus were isolated from Aedes spp. In ticks, the Langat virus was isolated from Ixodes spp. One of the major challenges in the diagnosis of FVs is the presence of sero-complexes as a result of cross-reactivity with one or more FV species. Subsequently, the distribution of specific FVs among humans and animals in a specific population is problematic to assess and often require comprehensive and thorough analyses. Molecular assays such as quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and digital droplet RT-PCR (ddRT-PCR) have been used for the differentiation of flavivirus infections to increase the accuracy of epidemiological data for disease surveillance, monitoring, and control. In situations where sero-complexes are common in FVs, even sensitive assays such as qRT-pCR can produce false positive results. In this write up, an overview of the various FV sero-complexes reported in Malaysia to date and the challenges faced in diagnosis of FV infections are presented.
Collapse
|
48
|
Smatti MK, Al Thani AA, Yassine HM. Viral-Induced Enhanced Disease Illness. Front Microbiol 2018; 9:2991. [PMID: 30568643 PMCID: PMC6290032 DOI: 10.3389/fmicb.2018.02991] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding immune responses to viral infections is crucial to progress in the quest for effective infection prevention and control. The host immunity involves various mechanisms to combat viral infections. Under certain circumstances, a viral infection or vaccination may result in a subverted immune system, which may lead to an exacerbated illness. Clinical evidence of enhanced illness by preexisting antibodies from vaccination, infection or maternal passive immunity is available for several viruses and is presumptively proposed for other viruses. Multiple mechanisms have been proposed to explain this phenomenon. It has been confirmed that certain infection- and/or vaccine-induced immunity could exacerbate viral infectivity in Fc receptor- or complement bearing cells- mediated mechanisms. Considering that antibody dependent enhancement (ADE) is a major obstacle in vaccine development, there are continues efforts to understand the underlying mechanisms through identification of the epitopes and antibodies responsible for disease enhancement or protection. This review discusses the recent findings on virally induced ADE, and highlights the potential mechanisms leading to this condition.
Collapse
Affiliation(s)
- Maria K Smatti
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
49
|
Baldwin WR, Livengood JA, Giebler HA, Stovall JL, Boroughs KL, Sonnberg S, Bohning KJ, Dietrich EA, Ong YT, Danh HK, Patel HK, Huang CYH, Dean HJ. Purified Inactivated Zika Vaccine Candidates Afford Protection against Lethal Challenge in Mice. Sci Rep 2018; 8:16509. [PMID: 30405178 PMCID: PMC6220238 DOI: 10.1038/s41598-018-34735-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 10/23/2018] [Indexed: 01/07/2023] Open
Abstract
In response to the 2016 global public health emergency of international concern announced by the World Health Organization surrounding Zika virus (ZIKV) outbreaks, we developed a purified inactivated Zika virus vaccine (PIZV) candidate from ZIKV strain PRVABC59, isolated during the outbreak in 2015. The virus isolate was plaque purified, creating six sub-isolated virus stocks, two of which were selected to generate PIZV candidates for preclinical immunogenicity and efficacy evaluation in mice. The alum-adjuvanted PIZV candidates were highly immunogenic in both CD-1 and AG129 mice after a 2-dose immunization. Further, AG129 mice receiving 2 doses of PIZV formulated with alum were fully protected against lethal ZIKV challenge and mouse immune sera elicited by the PIZV candidates were capable of neutralizing ZIKVs of both African and Asian genetic lineages in vitro. Additionally, passive immunization of naïve mice with ZIKV-immune serum showed strong positive correlation between neutralizing ZIKV antibody (NAb) titers and protection against lethal challenge. This study supported advancement of the PIZV candidate toward clinical development.
Collapse
Affiliation(s)
- Whitney R. Baldwin
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA ,0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Jill A. Livengood
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA
| | - Holli A. Giebler
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA ,0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Janae L. Stovall
- 0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Karen L. Boroughs
- 0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | | | - Kelly J. Bohning
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA
| | - Elizabeth A. Dietrich
- 0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Yee Tsuey Ong
- 0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Hoang K. Danh
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA
| | - Hetal K. Patel
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA
| | - Claire Y.-H. Huang
- 0000 0001 2163 0069grid.416738.fArboviral Diseases Branch, Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Hansi J. Dean
- 0000 0004 0447 7762grid.419849.9Takeda Vaccines Inc, Cambridge, MA USA
| |
Collapse
|
50
|
Shen WF, Galula JU, Liu JH, Liao MY, Huang CH, Wang YC, Wu HC, Liang JJ, Lin YL, Whitney MT, Chang GJJ, Chen SR, Wu SR, Chao DY. Epitope resurfacing on dengue virus-like particle vaccine preparation to induce broad neutralizing antibody. eLife 2018; 7:38970. [PMID: 30334522 PMCID: PMC6234032 DOI: 10.7554/elife.38970] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/18/2018] [Indexed: 12/25/2022] Open
Abstract
Dengue fever is caused by four different serotypes of dengue virus (DENV) which is the leading cause of worldwide arboviral diseases in humans. Virus-like particles (VLPs) containing flavivirus prM/E proteins have been demonstrated to be a potential vaccine candidate; however, the structure of dengue VLP is poorly understood. Herein VLP derived from DENV serotype-2 were engineered becoming highly matured (mD2VLP) and showed variable size distribution with diameter of ~31 nm forming the major population under cryo-electron microscopy examination. Furthermore, mD2VLP particles of 31 nm diameter possess a T = 1 icosahedral symmetry with a groove located within the E-protein dimers near the 2-fold vertices that exposed highly overlapping, cryptic neutralizing epitopes. Mice vaccinated with mD2VLP generated higher cross-reactive (CR) neutralization antibodies (NtAbs) and were fully protected against all 4 serotypes of DENV. Our results highlight the potential of ‘epitope-resurfaced’ mature-form D2VLPs in inducing quaternary structure-recognizing broad CR NtAbs to guide future dengue vaccine design.
Collapse
Affiliation(s)
- Wen-Fan Shen
- Microbial Genomics Ph.D. Program, National Chung Hsing University and Academia Sinica, Taichung City, Taiwan
| | - Jedhan Ucat Galula
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Jyung-Hurng Liu
- Institute of Genomics and Bioinformatics, College of Life Science, National Chung-Hsing University, Taichung City, Taiwan
| | - Mei-Ying Liao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Cheng-Hao Huang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Yu-Chun Wang
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| | - Han-Chung Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jian-Jong Liang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yi-Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Matthew T Whitney
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States
| | - Gwong-Jen J Chang
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States
| | - Sheng-Ren Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shang-Rung Wu
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Day-Yu Chao
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung City, Taiwan
| |
Collapse
|