1
|
Henriques P, Rosa A, Caldeira-Araújo H, Vigário AM. Mouse models as a tool to study asymptomatic DENV infections. Front Cell Infect Microbiol 2025; 15:1554090. [PMID: 40207056 PMCID: PMC11979173 DOI: 10.3389/fcimb.2025.1554090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/21/2025] [Indexed: 04/11/2025] Open
Abstract
Asymptomatic outcome accounts for most dengue virus infections and is likely to play an important role in maintaining virus circulation, contributing to its dissemination and shortening inter-epidemic periods. While dengue immunopathogenesis, investigation of potential therapeutics, and vaccine efficacy have been widely studied, only recently have inapparent infections begun to be comprehensively addressed as an integral and important part of the puzzle that is dengue infection. Animal models are one of the tools utilized to study dengue and, among these, mouse models have played an important role in understanding both dengue pathogenesis and the hosts' initial immune response. However, these models have mostly focused on untangling the drivers of disease severity ignoring asymptomatic dengue virus infections. In this mini-review, the authors propose to provide a concise overview of the current state-of-the-art of existing mouse models with potential use for studying asymptomatic dengue virus infections, elaborating on the pros and cons of the several models. Variations in experimental conditions, such as altering the viral load of the inoculum or employing different virus entry routes, especially in mice with partial or transient blockade of the type I interferon response, might be sufficient to obtain both symptomatic and asymptomatic viremic mice. This would enable the study of factors involved in asymptomatic dengue virus infections.
Collapse
Affiliation(s)
- Paulo Henriques
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Alexandra Rosa
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Centro de Química da Madeira (CQM), Universidade da Madeira, Funchal, Portugal
| | - Ana Margarida Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| |
Collapse
|
2
|
Baric TJ, Reneer ZB. Animal Models, Therapeutics, and Vaccine Approaches to Emerging and Re-Emerging Flaviviruses. Viruses 2024; 17:1. [PMID: 39861790 PMCID: PMC11769264 DOI: 10.3390/v17010001] [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: 11/13/2024] [Revised: 12/18/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
Flaviviruses are arthropod-borne viruses primarily transmitted through the mosquito Aedes aegypti or Culex genus of mosquitos. These viruses are predominantly found in tropical and subtropical regions of the world with their geographical spread predicted to increase as global temperatures continue to rise. These viruses cause a variety of diseases in humans with the most prevalent being caused by dengue, resulting in hemorrhagic fever and associated sequala. Current approaches for therapeutic control of flavivirus infections are limited, and despite recent advances, there are no approved drugs. Vaccines, available for a few circulating flaviviruses, still have limited potential for controlling contemporary and future outbreaks. Mouse models provide us with a valuable tool to test the effectiveness of drugs and vaccines, yet for many flaviviruses, well-established mouse models are lacking. In this review, we highlight the current state of flavivirus vaccines and therapeutics, as well as our current understanding of mouse models for various flaviviruses.
Collapse
Affiliation(s)
| | - Z. Beau Reneer
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3500, USA;
| |
Collapse
|
3
|
Yuya W, Yuansong Y, Susu L, Chen L, Yong W, Yining W, YouChun W, Changfa F. Progress and challenges in development of animal models for dengue virus infection. Emerg Microbes Infect 2024; 13:2404159. [PMID: 39312399 PMCID: PMC11423536 DOI: 10.1080/22221751.2024.2404159] [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: 05/27/2024] [Revised: 08/14/2024] [Accepted: 09/10/2024] [Indexed: 09/25/2024]
Abstract
ABSTRACTThe severity of the dengue epidemic is on the rise, with its geographic range had expanded to southern Europe by 2024. In this August, the WHO updated the pathogens that could spark the next pandemic, dengue virus was on the list. Vaccines and drugs serve as powerful tools for both preventing dengue infections and treating patients. Animal models play a pivotal role in vaccine development and drug screening. Available potential susceptible animals, including non-human primates, rodents, pigs, and tree shrews, have been extensively explored to establish animal models of dengue disease. Despite significant advancements, there are still notable limitations. Different animal models exhibit distinct constraining factors such as viraemia, host susceptibility, immune function of the host, clinical symptoms, ADE (antibody-dependent enhancement) phenomena, cytokine storm response to various serotypes and strain variations. Furthermore, despite extensive research on the dengue virus receptor in recent years, genetically modified animal models immunocompetent harbouring dengue virus susceptibility receptors have not yet been available. This work reviewed the research progress of dengue virus receptors and dengue animal models, suggesting that the development of genetically modified murine models expressing dengue virus functional receptors may hold a promise for future dengue disease research, especially for its vaccine development.
Collapse
Affiliation(s)
- Wang Yuya
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Yang Yuansong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Liu Susu
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Ling Chen
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
- College of Life Science school, Northwest University, Provincial Key Laboratory of Biotechnology of Shaanxi Province, Xi’an, People’s Republic of China
| | - Wu Yong
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang Yining
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| | - Wang YouChun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, People’s Republic of China
| | - Fan Changfa
- Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), Beijing, People’s Republic of China
| |
Collapse
|
4
|
Wu Z, He Y, Wang T, Wang M, Cheng A, Chen S. DENV and ZIKV infection: Species specificity and broad cell tropism. Virology 2024; 600:110276. [PMID: 39467358 DOI: 10.1016/j.virol.2024.110276] [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: 09/06/2024] [Revised: 10/14/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024]
Abstract
Nearly one-third of countries worldwide have reported cases of Dengue virus (DENV) and Zika virus (ZIKV) infections, highlighting the significant threat these viruses pose to global public health. As members of the Flavivirus genus within the Flaviviridae family, DENV and ZIKV have demonstrated the ability to infect a wide range of cell lines from multiple species in vitro. However, the range of susceptible animal models is notably limited, and field studies indicate that their capacity to infect host organisms is highly restricted, with a very narrow range of target cells in vivo. The virus's ability to hijack host cellular machinery plays a crucial role in determining its cellular and species specificity. In this review, we examine how DENV and ZIKV exploit host cells to facilitate their replication, offering new insights that could inform the development of antiviral drugs and therapeutic targets.
Collapse
Affiliation(s)
- Zhen Wu
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Yu He
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Tao Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Mingshu Wang
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China
| | - Anchun Cheng
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| | - Shun Chen
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China; Key Laboratory of Agricultural Bioinformatics of Ministry of Education, Sichuan Agricultural University, Chengdu, Sichuan, 611130, China.
| |
Collapse
|
5
|
Belmont L, Contreras M, Cartwright-Acar CH, Marceau CD, Agrawal A, Levoir LM, Lubow J, Goo L. Functional genomics screens reveal a role for TBC1D24 and SV2B in antibody-dependent enhancement of dengue virus infection. J Virol 2024; 98:e0158224. [PMID: 39377586 DOI: 10.1128/jvi.01582-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
Under some conditions, dengue virus (DENV) can hijack IgG antibodies to facilitate its uptake into target cells expressing Fc gamma receptors (FcgR)-a process known as antibody-dependent enhancement (ADE) of infection. Beyond a requirement for FcgR, host dependency factors for this unusual IgG-mediated infection route remain unknown. To identify cellular factors exclusively required for ADE, here, we performed CRISPR knockout (KO) screens in an in vitro system poorly permissive to infection in the absence of IgG antibodies. Validating our approach, a top hit was FcgRIIa, which facilitates the binding and internalization of IgG-bound DENV but is not required for canonical infection. Additionally, we identified host factors with no previously described role in DENV infection, including TBC1D24 and SV2B, which have known functions in regulated secretion. Using genetic knockout and trans-complemented cells, we validated a functional requirement for these host factors in ADE assays performed with monoclonal antibodies and polyclonal sera in multiple cell lines and using all four DENV serotypes. We show that knockout of TBC1D24 or SV2B impaired the binding of IgG-DENV complexes to cells without affecting FcgRIIa expression levels. Thus, we identify cellular factors beyond FcgR that promote efficient ADE of DENV infection. Our findings represent a first step toward advancing fundamental knowledge behind the biology of a non-canonical infection route implicated in disease.IMPORTANCEAntibodies can paradoxically enhance rather than inhibit dengue virus (DENV) infection in some cases. To advance knowledge of the functional requirements of antibody-dependent enhancement (ADE) of infection beyond existing descriptive studies, we performed a genome-scale CRISPR knockout (KO) screen in an optimized in vitro system permissive to efficient DENV infection only in the presence of IgG. In addition to FcgRIIa, a known receptor that facilitates IgG-mediated uptake of IgG-bound, but not naked DENV particles, our screens identified TBC1D24 and SV2B, cellular factors with no known role in DENV infection. We validated a functional role for TBC1D24 and SV2B in mediating ADE of all four DENV serotypes in different cell lines and using various antibodies. Thus, we identify cellular factors beyond Fc gamma receptors that promote ADE mechanisms. This study represents a first step toward advancing fundamental knowledge beyond a poorly understood non-canonical viral entry mechanism.
Collapse
Affiliation(s)
- Laura Belmont
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Maya Contreras
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | | | - Aditi Agrawal
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Lisa M Levoir
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jay Lubow
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Leslie Goo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
6
|
Sánchez JL, Salgado DM, Vega MR, Castro-Trujillo S, Narváez CF. Utility of the WHO dengue guidelines in pediatric immunological studies. J Trop Pediatr 2024; 70:fmae014. [PMID: 39002166 DOI: 10.1093/tropej/fmae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/15/2024]
Abstract
Dengue is a significant health problem due to the high burden of critical infections during outbreaks. In 1997, the World Health Organization (WHO) classified dengue as dengue fever (DF), dengue hemorrhagic fever (DHF), and dengue shock syndrome (DSS). It was revised in 2009 (updated in 2015), and the new guidelines recommended classifying patients as dengue without warning signs (DNS), dengue with warning signs (DWS), and severe dengue (SD). Although the utility of the revised 2009 classification for clinical studies is accepted, for immunological studies it needs to be clarified. We determined the usefulness of the 2009 classification for pediatric studies that analyze the circulating interleukin (IL)-6 and IL-8, two inflammatory cytokines. Plasma levels of IL-6 and IL-8 were evaluated in the acute and convalescent phases by flow cytometry in children with dengue classified using the 1997 and 2009 WHO guidelines. The plasma levels of IL-6 and IL-8 were elevated during the acute and decreased during convalescence, and both cytokines served as a good marker of acute dengue illness compared to convalescence. There were no differences in the plasma level of the evaluated cytokines among children with different clinical severity with any classification, except for the IL-8, which was higher in DWS than DNS. Based on the levels of IL-8, the 2009 classification identified DWS plus SD (hospital-treated children) compared to the DNS group [area under the curve (AUC): 0.7, p = 0.028]. These results support the utility of the revised 2009 (updated in 2015) classification in studies of immune markers in pediatric dengue.
Collapse
Affiliation(s)
- Jorge L Sánchez
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva 41001, Colombia
- Departamento de Pediatría, Universidad Surcolombiana, Hospital Universitario de Neiva, Neiva 41001, Colombia
| | - Doris M Salgado
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva 41001, Colombia
- Departamento de Pediatría, Universidad Surcolombiana, Hospital Universitario de Neiva, Neiva 41001, Colombia
| | - Martha Rocío Vega
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva 41001, Colombia
- Departamento de Pediatría, Universidad Surcolombiana, Hospital Universitario de Neiva, Neiva 41001, Colombia
| | - Sebastián Castro-Trujillo
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva 41001, Colombia
- Departamento de Pediatría, Universidad Surcolombiana, Hospital Universitario de Neiva, Neiva 41001, Colombia
| | - Carlos F Narváez
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva 41001, Colombia
- Departamento de Pediatría, Universidad Surcolombiana, Hospital Universitario de Neiva, Neiva 41001, Colombia
| |
Collapse
|
7
|
Belmont L, Contreras M, Cartwright-Acar CH, Marceau CD, Agrawal A, Levoir LM, Lubow J, Goo L. Functional genomics screens reveal a role for TBC1D24 and SV2B in antibody-dependent enhancement of dengue virus infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591029. [PMID: 38712102 PMCID: PMC11071485 DOI: 10.1101/2024.04.26.591029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Dengue virus (DENV) can hijack non-neutralizing IgG antibodies to facilitate its uptake into target cells expressing Fc gamma receptors (FcgR) - a process known as antibody-dependent enhancement (ADE) of infection. Beyond a requirement for FcgR, host dependency factors for this non-canonical infection route remain unknown. To identify cellular factors exclusively required for ADE, here, we performed CRISPR knockout screens in an in vitro system permissive to infection only in the presence of IgG antibodies. Validating our approach, a top hit was FcgRIIa, which facilitates binding and internalization of IgG-bound DENV but is not required for canonical infection. Additionally, we identified host factors with no previously described role in DENV infection, including TBC1D24 and SV2B, both of which have known functions in regulated secretion. Using genetic knockout and trans-complemented cells, we validated a functional requirement for these host factors in ADE assays performed with monoclonal antibodies and polyclonal sera in multiple cell lines and using all four DENV serotypes. We show that knockout of TBC1D24 or SV2B impaired binding of IgG-DENV complexes to cells without affecting FcgRIIa expression levels. Thus, we identify cellular factors beyond FcgR that are required for ADE of DENV infection. Our findings represent a first step towards advancing fundamental knowledge behind the biology of ADE that can ultimately be exploited to inform vaccination and therapeutic approaches.
Collapse
Affiliation(s)
- Laura Belmont
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Maya Contreras
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | | | - Aditi Agrawal
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Lisa M. Levoir
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jay Lubow
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Leslie Goo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
8
|
Wong MP, Juan EYW, Pahmeier F, Chelluri SS, Wang P, Castillo-Rojas B, Blanc SF, Biering SB, Vance RE, Harris E. The inflammasome pathway is activated by dengue virus non-structural protein 1 and is protective during dengue virus infection. PLoS Pathog 2024; 20:e1012167. [PMID: 38662771 DOI: 10.1371/journal.ppat.1012167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/07/2024] [Accepted: 04/01/2024] [Indexed: 05/07/2024] Open
Abstract
Dengue virus (DENV) is a medically important flavivirus causing an estimated 50-100 million dengue cases annually, some of whom progress to severe disease. DENV non-structural protein 1 (NS1) is secreted from infected cells and has been implicated as a major driver of dengue pathogenesis by inducing endothelial barrier dysfunction. However, less is known about how DENV NS1 interacts with immune cells and what role these interactions play. Here we report that DENV NS1 can trigger activation of inflammasomes, a family of cytosolic innate immune sensors that respond to infectious and noxious stimuli, in mouse and human macrophages. DENV NS1 induces the release of IL-1β in a caspase-1 dependent manner. Additionally, we find that DENV NS1-induced inflammasome activation is independent of the NLRP3, Pyrin, and AIM2 inflammasome pathways, but requires CD14. Intriguingly, DENV NS1-induced inflammasome activation does not induce pyroptosis and rapid cell death; instead, macrophages maintain cellular viability while releasing IL-1β. Lastly, we show that caspase-1/11-deficient, but not NLRP3-deficient, mice are more susceptible to lethal DENV infection. Together, these results indicate that the inflammasome pathway acts as a sensor of DENV NS1 and plays a protective role during infection.
Collapse
Affiliation(s)
- Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Evan Y W Juan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Sai S Chelluri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Phoebe Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, California, United States of America
- Howard Hughes Medical Institute, University of California, 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
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, California, United States of America
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, California, United States of America
| |
Collapse
|
9
|
Siddqui G, Vishwakarma P, Saxena S, Kumar V, Bajpai S, Kumar A, Kumar S, Khatri R, Kaur J, Bhattacharya S, Ahmed S, Syed GH, Kumar Y, Samal S. Aged AG129 mice support the generation of highly virulent novel mouse-adapted DENV (1-4) viruses exhibiting neuropathogenesis and high lethality. Virus Res 2024; 341:199331. [PMID: 38280436 PMCID: PMC10846402 DOI: 10.1016/j.virusres.2024.199331] [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: 09/25/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 01/29/2024]
Abstract
Dengue virus infection in humans ranges from asymptomatic infection to severe infection, with ∼2.5 % overall disease fatality rate. Evidence of neurological manifestations is seen in the severe form of the disease, which might be due to the direct invasion of the viruses into the CNS system but is poorly understood. In this study, we demonstrated that the aged AG129 mice are highly susceptible to dengue serotypes 1-4, and following the adaptation, this resulted in the generation of neurovirulent strains that showed enhanced replication, aggravated disease severity, increased neuropathogenesis, and high lethality in both adult and aged AG129 mice. The infected mice had endothelial dysfunction, elicited pro-inflammatory cytokine responses, and exhibited 100 % mortality. Further analysis revealed that aged-adapted DENV strains induced measurable alterations in TLR expression in the aged mice as compared to the adult mice. In addition, metabolomics analysis of the serum samples from the infected adult mice revealed dysregulation of 18 metabolites and upregulation of 6-keto-prostaglandin F1 alpha, phosphocreatine, and taurocholic acid. These metabolites may serve as key biomarkers to decipher and comprehend the severity of dengue-associated severe neuro-pathogenesis.
Collapse
Affiliation(s)
- Gazala Siddqui
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Preeti Vishwakarma
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Shikha Saxena
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Varun Kumar
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Sneh Bajpai
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Amit Kumar
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Satish Kumar
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Ritika Khatri
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Jaskaran Kaur
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Sankar Bhattacharya
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Shubbir Ahmed
- All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | | | - Yashwant Kumar
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India
| | - Sweety Samal
- Influenza and Respiratory Virus Laboratory, Centre for Virus Research, Therapeutics and Vaccines, Translational Health Science and Technology Institute, NCR-Biotech Science Cluster, 3rd Milestossne, Faridabad-Gurgaon Expressway, Faridabad, Haryana 121001, India.
| |
Collapse
|
10
|
Caetano CCS, Azamor T, Meyer NM, Onwubueke C, Calabrese CM, Calabrese LH, Visperas A, Piuzzi NS, Husni ME, Foo SS, Chen W. Mechanistic insights into bone remodelling dysregulation by human viral pathogens. Nat Microbiol 2024; 9:322-335. [PMID: 38316931 PMCID: PMC11045166 DOI: 10.1038/s41564-023-01586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 12/12/2023] [Indexed: 02/07/2024]
Abstract
Bone-related diseases (osteopathologies) associated with human virus infections have increased around the globe. Recent findings have highlighted the intricate interplay between viral infection, the host immune system and the bone remodelling process. Viral infections can disrupt bone homeostasis, contributing to conditions such as arthritis and soft tissue calcifications. Osteopathologies can occur after arbovirus infections such as chikungunya virus, dengue virus and Zika virus, as well as respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 and enteroviruses such as Coxsackievirus B. Here we explore how human viruses dysregulate bone homeostasis, detailing viral factors, molecular mechanisms, host immune response changes and bone remodelling that ultimately result in osteopathologies. We highlight model systems and technologies to advance mechanistic understanding of viral-mediated bone alterations. Finally, we propose potential prophylactic and therapeutic strategies, introduce 'osteovirology' as a research field highlighting the underestimated roles of viruses in bone-related diseases, and discuss research avenues for further investigation.
Collapse
Affiliation(s)
- Camila C S Caetano
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tamiris Azamor
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nikki M Meyer
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Chineme Onwubueke
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Cassandra M Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Leonard H Calabrese
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Anabelle Visperas
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Nicolas S Piuzzi
- Department of Orthopedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - M Elaine Husni
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Suan-Sin Foo
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| | - Weiqiang Chen
- Infection Biology Program, Global Center for Pathogen Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
11
|
Marano JM, Weger-Lucarelli J. Preexisting inter-serotype immunity drives antigenic evolution of dengue virus serotype 2. Virology 2024; 590:109951. [PMID: 38096749 PMCID: PMC10855010 DOI: 10.1016/j.virol.2023.109951] [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: 09/19/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Dengue virus (DENV) infects roughly 400 million people annually, causing febrile and hemorrhagic disease. While preexisting inter-serotype immunity (PISI) provides transient protection, it may drive severe disease over time. PISI's impact on virus evolution, however, is less understood. Retrospective epidemiological analyses suggest that PISI may drive DENV evolution. Using in vitro directed evolution, we explored how DENV2 evolves in the presence of DENV3/4 convalescent serum. Two post-passaging mutations (E-I6M and E-N203D) were then studied for fitness effects in mammalian and insect hosts and immune escape. E-I6M resisted neutralization, altered fitness in mammalian cell culture models, and had no effect in Aedes albopictus mosquitoes. E-N203D showed no change in neutralization sensitivity, reduced fitness in a DENV-naïve epithelial model, and no effects in the other models. These results align with surveillance data, where E-I6M emerged and disappeared, while E-203D and E-203 N cocirculate, thus suggesting that PISI can drive DENV evolution.
Collapse
Affiliation(s)
- Jeffrey M Marano
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Roanoke, VA, United States; Department of Biomedical Sciences and Pathobiology, Virginia Tech, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States; Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, United States.
| |
Collapse
|
12
|
Malavige GN, Sjö P, Singh K, Piedagnel JM, Mowbray C, Estani S, Lim SCL, Siquierra AM, Ogg GS, Fraisse L, Ribeiro I. Facing the escalating burden of dengue: Challenges and perspectives. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0002598. [PMID: 38100392 PMCID: PMC10723676 DOI: 10.1371/journal.pgph.0002598] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
Dengue is the most rapidly emerging mosquito-borne infection and, due to climate change and unplanned urbanization, it is predicted that the global burden of dengue will rise further as the infection spreads to new geographical locations. Dengue-endemic countries are often unable to cope with such increases, with health care facilities becoming overwhelmed during each dengue season. Furthermore, although dengue has been predominantly a childhood illness in the past, it currently mostly affects adults in many countries, with higher incidence of severe disease and mortality rates in pregnant women and in those with comorbidities. As there is currently no specific treatment for dengue and no early biomarker to identify those who will progress to develop vascular leakage, all individuals with dengue are closely monitored in case they need fluid management. Furthermore, diagnosing patients with acute dengue is challenging due to the similarity of clinical symptoms during early illness and poor sensitivity and specificity of point-of-care diagnostic tests. Novel vector control methods, such as the release of Wolbachia-infected mosquitoes, have shown promising results by reducing vector density and dengue incidence in clinical trial settings. A new dengue vaccine, TAK-003, had an efficacy of 61.2% against virologically confirmed dengue, 84.1% efficacy against hospitalizations and a 70% efficacy against development of dengue haemorrhagic fever (DHF) at 54 months. While vaccines and mosquito control methods are welcome, they alone are unlikely to fully reduce the burden of dengue, and a treatment for dengue is therefore essential. Several novel antiviral drugs are currently being evaluated along with drugs that inhibit host mediators, such as mast cell products. Although viral proteins such as NS1 contribute to the vascular leak observed in severe dengue, the host immune response to the viral infection also plays a significant role in progression to severe disease. There is an urgent need to discover safe and effective treatments for dengue to prevent disease progression.
Collapse
Affiliation(s)
- Gathsaurie Neelika Malavige
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Peter Sjö
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Kavita Singh
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | | | - Charles Mowbray
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Sergio Estani
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | | | | | - Graham S. Ogg
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Laurent Fraisse
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Isabela Ribeiro
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| |
Collapse
|
13
|
Henriques P, Rosa A, Caldeira-Araújo H, Soares P, Vigário AM. Flying under the radar - impact and factors influencing asymptomatic DENV infections. Front Cell Infect Microbiol 2023; 13:1284651. [PMID: 38076464 PMCID: PMC10704250 DOI: 10.3389/fcimb.2023.1284651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
The clinical outcome of DENV and other Flaviviruses infections represents a spectrum of severity that ranges from mild manifestations to severe disease, which can ultimately lead to death. Nonetheless, most of these infections result in an asymptomatic outcome that may play an important role in the persistent circulation of these viruses. Also, although little is known about the mechanisms that lead to these asymptomatic infections, they are likely the result of a complex interplay between viral and host factors. Specific characteristics of the infecting viral strain, such as its replicating efficiency, coupled with host factors, like gene expression of key molecules involved in the immune response or in the protection against disease, are among crucial factors to study. This review revisits recent data on factors that may contribute to the asymptomatic outcome of the world's widespread DENV, highlighting the importance of silent infections in the transmission of this pathogen and the immune status of the host.
Collapse
Affiliation(s)
- Paulo Henriques
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Alexandra Rosa
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
| | - Helena Caldeira-Araújo
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- CQM-Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Pedro Soares
- Department of Biology, CBMA (Centre of Molecular and Environmental Biology), Braga, Portugal
- Department of Biology, Institute of Science and Innovation for Bio-Sustainability (IB-S), University of Minho, Braga, Portugal
| | - Ana Margarida Vigário
- Projecto Medicina, Faculdade de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
14
|
Kuhn RJ, Barrett ADT, Desilva AM, Harris E, Kramer LD, Montgomery RR, Pierson TC, Sette A, Diamond MS. A Prototype-Pathogen Approach for the Development of Flavivirus Countermeasures. J Infect Dis 2023; 228:S398-S413. [PMID: 37849402 PMCID: PMC10582523 DOI: 10.1093/infdis/jiad193] [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: 02/22/2023] [Accepted: 05/28/2023] [Indexed: 10/19/2023] Open
Abstract
Flaviviruses are a genus within the Flaviviridae family of positive-strand RNA viruses and are transmitted principally through mosquito and tick vectors. These viruses are responsible for hundreds of millions of human infections worldwide per year that result in a range of illnesses from self-limiting febrile syndromes to severe neurotropic and viscerotropic diseases and, in some cases, death. A vaccine against the prototype flavivirus, yellow fever virus, has been deployed for 85 years and is highly effective. While vaccines against some medically important flaviviruses are available, others have proven challenging to develop. The emergence and spread of flaviviruses, including dengue virus and Zika virus, demonstrate their pandemic potential. This review highlights the gaps in knowledge that need to be addressed to allow for the rapid development of vaccines against emerging flaviviruses in the future.
Collapse
Affiliation(s)
- Richard J Kuhn
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana, USA
- Purdue Institute of Inflammation, Immunology, and Infectious Disease, Purdue University, West Lafayette, Indiana, USA
| | - Alan D T Barrett
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
| | - Aravinda M Desilva
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California Berkeley, Berkeley, California, USA
| | - Laura D Kramer
- School of Public Health, State University of New York at Albany, Albany, New York, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Theodore C Pierson
- Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, University of California in San Diego, San Diego, California, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St Louis, Missouri, USA
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
15
|
Lubow J, Levoir LM, Ralph DK, Belmont L, Contreras M, Cartwright-Acar CH, Kikawa C, Kannan S, Davidson E, Duran V, Rebellon-Sanchez DE, Sanz AM, Rosso F, Doranz BJ, Einav S, Matsen IV FA, Goo L. Single B cell transcriptomics identifies multiple isotypes of broadly neutralizing antibodies against flaviviruses. PLoS Pathog 2023; 19:e1011722. [PMID: 37812640 PMCID: PMC10586629 DOI: 10.1371/journal.ppat.1011722] [Citation(s) in RCA: 2] [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: 10/19/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023] Open
Abstract
Sequential dengue virus (DENV) infections often generate neutralizing antibodies against all four DENV serotypes and sometimes, Zika virus. Characterizing cross-flavivirus broadly neutralizing antibody (bnAb) responses can inform countermeasures that avoid enhancement of infection associated with non-neutralizing antibodies. Here, we used single cell transcriptomics to mine the bnAb repertoire following repeated DENV infections. We identified several new bnAbs with comparable or superior breadth and potency to known bnAbs, and with distinct recognition determinants. Unlike all known flavivirus bnAbs, which are IgG1, one newly identified cross-flavivirus bnAb (F25.S02) was derived from IgA1. Both IgG1 and IgA1 versions of F25.S02 and known bnAbs displayed neutralizing activity, but only IgG1 enhanced infection in monocytes expressing IgG and IgA Fc receptors. Moreover, IgG-mediated enhancement of infection was inhibited by IgA1 versions of bnAbs. We demonstrate a role for IgA in flavivirus infection and immunity with implications for vaccine and therapeutic strategies.
Collapse
Affiliation(s)
- Jay Lubow
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Lisa M. Levoir
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Duncan K. Ralph
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Laura Belmont
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, United States of America
| | - Maya Contreras
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Catiana H. Cartwright-Acar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Caroline Kikawa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- Medical Scientist Training Program, University of Washington, Seattle, Washington, United States of America
| | - Shruthi Kannan
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Edgar Davidson
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Veronica Duran
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | | | - Ana M. Sanz
- Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
| | - Fernando Rosso
- Clinical Research Center, Fundación Valle del Lili, Cali, Colombia
- Department of Internal Medicine, Division of Infectious Diseases, Fundación Valle del Lili, Cali, Colombia
| | - Benjamin J. Doranz
- Integral Molecular, Inc., Philadelphia, Pennsylvania, United States of America
| | - Shirit Einav
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Frederick A. Matsen IV
- Computational Biology Program, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
- Department of Genome Sciences, University of Washington, Seattle, Washington, United States of America
- Department of Statistics, University of Washington, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Seattle, Washington, United States of America
| | - Leslie Goo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
16
|
Wong MP, Juan EYW, Chelluri SS, Wang P, Pahmeier F, Castillo-Rojas B, Blanc SF, Biering SB, Vance RE, Harris E. The Inflammasome Pathway is Activated by Dengue Virus Non-structural Protein 1 and is Protective During Dengue Virus Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558875. [PMID: 37790301 PMCID: PMC10543007 DOI: 10.1101/2023.09.21.558875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dengue virus (DENV) is a medically important flavivirus causing an estimated 50-100 million dengue cases annually, some of whom progress to severe disease. DENV non-structural protein 1 (NS1) is secreted from infected cells and has been implicated as a major driver of dengue pathogenesis by inducing endothelial barrier dysfunction. However, less is known about how DENV NS1 interacts with immune cells and what role these interactions play. Here we report that DENV NS1 can trigger activation of inflammasomes, a family of cytosolic innate immune sensors that respond to infectious and noxious stimuli, in mouse and human macrophages. DENV NS1 induces the release of IL-1β in a caspase-1 dependent manner. Additionally, we find that DENV NS1-induced inflammasome activation is independent of the NLRP3, Pyrin, and AIM2 inflammasome pathways, but requires CD14. Intriguingly, DENV NS1-induced inflammasome activation does not induce pyroptosis and rapid cell death; instead, macrophages maintain cellular viability while releasing IL-1β. Lastly, we show that caspase-1/11-deficient, but not NLRP3-deficient, mice are more susceptible to lethal DENV infection. Together, these results indicate that the inflammasome pathway acts as a sensor of DENV NS1 and plays a protective role during infection.
Collapse
Affiliation(s)
- Marcus P Wong
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Evan Y W Juan
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sai S Chelluri
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Phoebe Wang
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Felix Pahmeier
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Bryan Castillo-Rojas
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Sophie F Blanc
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Russell E Vance
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, California, USA
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
17
|
Nemirov K, Authié P, Souque P, Moncoq F, Noirat A, Blanc C, Bourgine M, Majlessi L, Charneau P. Preclinical proof of concept of a tetravalent lentiviral T-cell vaccine against dengue viruses. Front Immunol 2023; 14:1208041. [PMID: 37654495 PMCID: PMC10466046 DOI: 10.3389/fimmu.2023.1208041] [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: 04/18/2023] [Accepted: 07/17/2023] [Indexed: 09/02/2023] Open
Abstract
Dengue virus (DENV) is responsible for approximately 100 million cases of dengue fever annually, including severe forms such as hemorrhagic dengue and dengue shock syndrome. Despite intensive vaccine research and development spanning several decades, a universally accepted and approved vaccine against dengue fever has not yet been developed. The major challenge associated with the development of such a vaccine is that it should induce simultaneous and equal protection against the four DENV serotypes, because past infection with one serotype may greatly increase the severity of secondary infection with a distinct serotype, a phenomenon known as antibody-dependent enhancement (ADE). Using a lentiviral vector platform that is particularly suitable for the induction of cellular immune responses, we designed a tetravalent T-cell vaccine candidate against DENV ("LV-DEN"). This vaccine candidate has a strong CD8+ T-cell immunogenicity against the targeted non-structural DENV proteins, without inducing antibody response against surface antigens. Evaluation of its protective potential in the preclinical flavivirus infection model, i.e., mice knockout for the receptor to the type I IFN, demonstrated its significant protective effect against four distinct DENV serotypes, based on reduced weight loss, viremia, and viral loads in peripheral organs of the challenged mice. These results provide proof of concept for the use of lentiviral vectors for the development of efficient polyvalent T-cell vaccine candidates against all DENV serotypes.
Collapse
Affiliation(s)
- Kirill Nemirov
- Pasteur-TheraVectys Joint Lab, Institut Pasteur, Université de Paris, Virology Department, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
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: 1.5] [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
|
19
|
Ooi EE, Kalimuddin S. Insights into dengue immunity from vaccine trials. Sci Transl Med 2023; 15:eadh3067. [PMID: 37437017 DOI: 10.1126/scitranslmed.adh3067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
The quest for an effective dengue vaccine has culminated in two approved vaccines and another that has completed phase 3 clinical trials. However, shortcomings exist in each, suggesting that the knowledge on dengue immunity used to develop these vaccines was incomplete. Vaccine trial findings could refine our understanding of dengue immunity, because these are experimentally derived, placebo-controlled data. Results from these trials suggest that neutralizing antibody titers alone are insufficient to inform protection against symptomatic infection, implicating a role for cellular immunity in protection. These findings have relevance for both future dengue vaccine development and application of current vaccines for maximal public health benefit.
Collapse
Affiliation(s)
- Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Centre, SingHealth Duke-NUS Academic Medical Centre, Singapore 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore 117549, Singapore
| | - Shirin Kalimuddin
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Infectious Diseases, Singapore General Hospital, Singapore 169856, Singapore
| |
Collapse
|
20
|
Lubow J, Levoir LM, Ralph DK, Belmont L, Contreras M, Cartwright-Acar CH, Kikawa C, Kannan S, Davidson E, Doranz BJ, Duran V, Sanchez DE, Sanz AM, Rosso F, Einav S, Matsen FA, Goo L. Single B cell transcriptomics identifies multiple isotypes of broadly neutralizing antibodies against flaviviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.09.536175. [PMID: 37090561 PMCID: PMC10120628 DOI: 10.1101/2023.04.09.536175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Sequential dengue virus (DENV) infections often generate neutralizing antibodies against all four DENV serotypes and sometimes, Zika virus. Characterizing cross-flavivirus broadly neutralizing antibody (bnAb) responses can inform countermeasure strategies that avoid infection enhancement associated with non-neutralizing antibodies. Here, we used single cell transcriptomics to mine the bnAb repertoire following secondary DENV infection. We identified several new bnAbs with comparable or superior breadth and potency to known bnAbs, and with distinct recognition determinants. Unlike all known flavivirus bnAbs, which are IgG1, one newly identified cross-flavivirus bnAb (F25.S02) was derived from IgA1. Both IgG1 and IgA1 versions of F25.S02 and known bnAbs displayed neutralizing activity, but only IgG1 enhanced infection in monocytes expressing IgG and IgA Fc receptors. Moreover, IgG-mediated enhancement of infection was inhibited by IgA1 versions of bnAbs. We demonstrate a role for IgA in flavivirus infection and immunity with implications for vaccine and therapeutic strategies.
Collapse
|
21
|
Wilken L, Stelz S, Prajeeth CK, Rimmelzwaan GF. Transient Blockade of Type I Interferon Signalling Promotes Replication of Dengue Virus Strain D2Y98P in Adult Wild-Type Mice. Viruses 2023; 15:v15040814. [PMID: 37112795 PMCID: PMC10142689 DOI: 10.3390/v15040814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Dengue virus serotypes 1 to 4 (DENV1-4) place nearly half the global population at risk of infection and the licenced tetravalent dengue vaccine fails to protect individuals who have not previously been exposed to DENV. The development of intervention strategies had long been hampered by the lack of a suitable small animal model. DENV does not replicate in wild-type mice due to its inability to antagonise the mouse type I interferon (IFN) response. Mice deficient in type I IFN signalling (Ifnar1-/- mice) are highly susceptible to DENV infection, but their immunocompromised status makes it difficult to interpret immune responses elicited by experimental vaccines. To develop an alternative mouse model for vaccine testing, we treated adult wild-type mice with MAR1-5A3-an IFNAR1-blocking, non-cell-depleting antibody-prior to infection with the DENV2 strain D2Y98P. This approach would allow for vaccination of immunocompetent mice and subsequent inhibition of type I IFN signalling prior to challenge infection. While Ifnar1-/- mice quickly succumbed to infection, MAR1-5A3-treated mice did not show any signs of illness but eventually seroconverted. Infectious virus was recovered from the sera and visceral organs of Ifnar1-/- mice, but not from those of mice treated with MAR1-5A3. However, high levels of viral RNA were detected in the samples of MAR1-5A3-treated mice, indicating productive viral replication and dissemination. This transiently immunocompromised mouse model of DENV2 infection will aid the pre-clinical assessment of next-generation vaccines as well as novel antiviral treatments.
Collapse
Affiliation(s)
- Lucas Wilken
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Sonja Stelz
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Chittappen Kandiyil Prajeeth
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| | - Guus F Rimmelzwaan
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine (TiHo), 30559 Hannover, Germany
| |
Collapse
|
22
|
Shi LZ, Chen X, Cao HH, Tian CY, Zou LF, Yu JH, Lu ZB, Zhao W, Liu JS, Yu LZ. N-Butanol Extract of Glycyrrhizae Radix et Rhizoma Inhibits Dengue Virus through Targeting Envelope Protein. Pharmaceuticals (Basel) 2023; 16:263. [PMID: 37259408 PMCID: PMC9962983 DOI: 10.3390/ph16020263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/13/2023] [Accepted: 01/23/2023] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND At present, about half of the world's population is at risk of being infected with dengue virus (DENV). However, there are no specific drugs to prevent or treat DENV infection. Glycyrrhizae Radix et Rhizome, a well-known traditional Chinese medicine, performs multiple pharmacological activities, including exerting antiviral effects. The aim of this study was to investigate the anti-DENV effects of n-butanol extract from Glycyrrhizae Radix et Rhizome (GRE). METHODS Compounds analysis of GRE was conducted via ultra-performance liquid chromatography/tandem mass spectrometry (UHPLC-MS/MS). The antiviral activities of GRE were determined by the CCK-8 assay, plaque assay, qRT-PCR, Western blotting, and the immunofluorescence assay. The DENV-infected suckling mice model was constructed to explore the antiviral effects of GRE in vivo. RESULTS Four components in GRE were analyzed by UHPLC-MS/MS, including glycyrrhizic acid, glycyrrhetnic acid, liquiritigenin, and isoliquiritigenin. GRE inhibited the attachment process of the virus replication cycle and reduced the expression of the E protein in cell models. In the in vivo study, GRE significantly relieved clinical symptoms and prolong survival duration. GRE also significantly decreased viremia, reduced the viral load in multiple organs, and inhibited the release of pro-inflammatory cytokines in DENV-infected suckling mice. CONCLUSIONS GRE exhibited significant inhibitory activities in the adsorption stage of the DENV-2 replication cycle by targeting the envelope protein. Thus, GRE might be a promising candidate for the treatment of DENV infection.
Collapse
Affiliation(s)
- Ling-Zhu Shi
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xi Chen
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hui-Hui Cao
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chun-Yang Tian
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Li-Fang Zou
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Jian-Hai Yu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zi-Bin Lu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Jun-Shan Liu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Lin-Zhong Yu
- Third Level Research Laboratory of State Administration of Traditional Chinese Medicine, Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
23
|
Liu X, Liu Y, Wu H, He Z, Li Z, Qin Z, Yu J, Zhu L, Wu Q, Xiao W, Shen C, Wan C, Zhang B, Zhao W. Murine diabetic models for dengue virus infection. J Med Virol 2022; 94:5943-5953. [PMID: 36000451 DOI: 10.1002/jmv.28088] [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: 06/20/2022] [Revised: 07/26/2022] [Accepted: 08/16/2022] [Indexed: 01/06/2023]
Abstract
Dengue virus (DENV) is a critical public health concern in tropical and subtropical regions worldwide. Thus, immunocompetent murine models of DENV infection with robust viremia are required for vaccine studies. Diabetes is highly prevalent worldwide, making it frequent comorbidity in patients with dengue fever. Therefore, murine models are needed to understand viral pathogenesis and disease progression. Acquired-induced and inherently diabetic C57BL/6 and db/db mice were inoculated with DENV-3 via the tail vein. After infection, both the diabetic C57BL/6 and db/db mice showed obvious weight loss with clinical manifestations. Quantitative reverse-transcription polymerase chain reaction revealed robust and replicable viremia in the two types of diabetic mice. Immunohistochemical detection showed persistent DENV-3 infection in the liver. Enzyme-linked immunosorbent assay for cytokine detection revealed that diabetic mice showed more severe inflammatory responses than did nondiabetic mice, and significant histological alterations were observed in diabetic mice. Thus, the diabetic mice were more susceptible to DENV infection than the nondiabetic mice. Taken together, we established two types of immunocompetent diabetic mice for DENV infection, which can be used to further study the mechanisms of dengue pathogenesis in diabetes and to develop antiviral pharmaceuticals and treatments.
Collapse
Affiliation(s)
- Xuling Liu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Yingfang Liu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Hao Wu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Zihan He
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhuoyun Li
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhiran Qin
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Jianhai Yu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Li Zhu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Qinghua Wu
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Weiwei Xiao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Chenguang Shen
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Chengsong Wan
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Bao Zhang
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| | - Wei Zhao
- BSL-3 Laboratory (Guangdong), Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
24
|
Engineering Modified mRNA-Based Vaccine against Dengue Virus Using Computational and Reverse Vaccinology Approaches. Int J Mol Sci 2022; 23:ijms232213911. [PMID: 36430387 PMCID: PMC9698390 DOI: 10.3390/ijms232213911] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/07/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Dengue virus belonging to the family Flaviviridae and its four serotypes are responsible for dengue infections, which extend over 60 countries in tropical and subtropical areas of the world including Pakistan. During the ongoing dengue outbreak in Pakistan (2022), over 30,000 cases have been reported, and over 70 lives have been lost. The only commercialized vaccine against DENV, Dengvaxia, cannot be administered as a prophylactic measure to cure this infection due to various complications. Using machine learning and reverse vaccinology approaches, this study was designed to develop a tetravalent modified nucleotide mRNA vaccine using NS1, prM, and EIII sequences of dengue virus from Pakistani isolates. Based on high antigenicity, non-allergenicity, and toxicity profiling, B-cell epitope, cytotoxic T lymphocyte (CTL), and helper T lymphocyte (HTL) putative vaccine targets were predicted. Molecular docking confirmed favorable interactions between T-cell epitopes and their respective HLA alleles, while normal mode analysis validated high-affinity interactions of vaccine proteins with immune receptors. In silico immune simulations confirmed adequate immune responses to eliminate the antigen and generate memory. Codon optimization, physicochemical features, nucleotide modifications, and suitable vector availability further ensured better antigen expression and adaptive immune responses. We predict that this vaccine construct may prove to be a good vaccinal candidate against dengue virus in vitro as well.
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Dengue vaccine development is a high public health priority. To date, no dengue vaccine is in widespread use. Here we review the challenges in dengue development and the latest results for the second-generation dengue vaccines. RECENT FINDINGS The biggest hurdle is the immunological interaction between the four antigenically distinct dengue serotypes. The advantages of second-generation dengue vaccines are the inclusion of nonstructural proteins of the dengue backbone and a more convenient dosing with reduced numbers of doses needed. SUMMARY Although dengue-primed individuals can already benefit from vaccination with the first licensed dengue vaccine CYD-TDV, the public health need for the dengue-naive population has not yet been met. The urgent need remains to identify correlates of both protection and enhancement; until such correlates have been identified, all second-generation dengue vaccines still need to go through full phase 3 trials. The 5-year efficacy and safety data for both second-generation dengue vaccines are imminent.
Collapse
Affiliation(s)
- Annelies Wilder-Smith
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
26
|
Abstract
Zika virus (ZIKV) is unusual among flaviviruses in its ability to spread between humans through sexual contact, as well as by mosquitoes. Sexual transmission has the potential to change the epidemiology and geographic range of ZIKV compared to mosquito-borne transmission and potentially could produce distinct clinical manifestations, so it is important to understand the host mechanisms that control susceptibility to sexually transmitted ZIKV. ZIKV replicates poorly in wild-type mice following subcutaneous inoculation, so most ZIKV pathogenesis studies use mice lacking type I interferon (IFN-αβ) signaling (e.g., Ifnar1-/-). We found that wild-type mice support ZIKV replication following intravaginal infection, consistent with prior studies, although the infection remained localized to the lower female reproductive tract. Vaginal ZIKV infection required a high-progesterone state (pregnancy or pretreatment with depot medroxyprogesterone acetate [DMPA]) even in Ifnar1-/- mice that otherwise are highly susceptible to ZIKV infection. Progesterone-mediated susceptibility did not appear to result from a compromised epithelial barrier, blunted antiviral gene induction, or changes in vaginal leukocyte populations, leaving open the mechanism by which progesterone confers susceptibility to vaginal ZIKV infection. DMPA treatment is a key component of mouse vaginal infection models for herpes simplex virus and Chlamydia, but the mechanisms by which DMPA increases susceptibility to those pathogens also remain poorly defined. Understanding how progesterone mediates susceptibility to ZIKV vaginal infection may provide insights into host mechanisms influencing susceptibility to diverse sexually transmitted pathogens. IMPORTANCE Zika virus (ZIKV) is transmitted by mosquitoes, similar to other flaviviruses. However, ZIKV is unusual among flaviviruses in its ability also to spread through sexual transmission. We found that ZIKV was able to replicate in the vaginas of wild-type mice, even though these mice do not support ZIKV replication by other routes, suggesting that the vagina is particularly susceptible to ZIKV infection. Vaginal susceptibility was dependent on a high-progesterone state, which is a common feature of mouse vaginal infection models for other pathogens, through mechanisms that have remained poorly defined. Understanding how progesterone mediates susceptibility to ZIKV vaginal infection may provide insights into host mechanisms that influence susceptibility to diverse sexually transmitted pathogens.
Collapse
|
27
|
Altamish M, Khan M, Baig MS, Pathak B, Rani V, Akhtar J, Khan AA, Ahmad S, Krishnan A. Therapeutic Potential of Medicinal Plants against Dengue Infection: A Mechanistic Viewpoint. ACS OMEGA 2022; 7:24048-24065. [PMID: 35874231 PMCID: PMC9301714 DOI: 10.1021/acsomega.2c00625] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Dengue is a tropical disease caused by the Dengue virus (DENV), a positive-sense, single stranded RNA virus of the family Flaviviridae, which is transmitted by Aedes mosquitoes. The occurrence of dengue has grown dramatically around the globe in recent decades, and it is rapidly becoming a global burden. Furthermore, all four DENV serotypes cocirculate and create a problematic hyperendemic situation. Characteristic symptoms range from being asymptomatic, dengue fever to life-threatening complications such as hemorrhagic fever and shock. Apart from the inherent virulence of the virus strain, a dysregulated host immune response makes the condition worse. Currently, there is no highly recommended vaccine or therapeutic agent against dengue. With the advent of virus strains resistant to antiviral agents, there is a constant need for new therapies to be developed. Since time immemorial, human civilization has utilized plants in traditional medicine to treat various diseases, including infectious viral diseases. With the advancement in molecular biology, cell biology techniques, and bioinformatics, recent studies have tried to provide scientific evidence and determine the mechanism of anti-dengue activity of various plant extracts and plant-derived agents. The current Review consolidates the studies on the last 20 years of in vitro and in vivo experiments on the ethnomedicinal plants used against the dengue virus. Several active phytoconstituents like quercetin, castanospermine, α-mangostin, schisandrin-A, hirsutin have been found to be promising to inhibition of all the four DENV serotypes. However, novel therapeutics need to be reassessed in relevant cells using high-throughput techniques. Further, in vivo dose optimization for the immunomodulatory and antiviral activity should be examined on a vast sample size. Such a Review should help take the knowledge forward, validate it, and use medicinal plants in different combinations targeting multiple stages of virus infection for more effective multipronged therapy against dengue infection.
Collapse
Affiliation(s)
- Mohammad Altamish
- Department
of Pharmacology School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Muzayyana Khan
- Bioactive
Natural Product Laboratory, School of Pharmaceutical Education and
Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mirza Sarwar Baig
- Department
of Molecular Medicine, School of Interdisciplinary Sciences &
Technology, Jamia Hamdard, New Delhi-110062 India
| | - Bharti Pathak
- Department
of Molecular Medicine, School of Interdisciplinary Sciences &
Technology, Jamia Hamdard, New Delhi-110062 India
| | - Veena Rani
- Department
of SciencesIndira Gandhi National Open University
(IGNOU), New Delhi, 110068, India
| | - Jamal Akhtar
- Central
Council for Research in Unani Medicine, Ministry of AYUSH, Government
of India, New Delhi, 110058, India
| | - A. Ali Khan
- Central
Council for Research in Unani Medicine, Ministry of AYUSH, Government
of India, New Delhi, 110058, India
| | - Sayeed Ahmad
- Bioactive
Natural Product Laboratory, School of Pharmaceutical Education and
Research, Jamia Hamdard, New Delhi, 110062, India
| | - Anuja Krishnan
- Department
of Molecular Medicine, School of Interdisciplinary Sciences &
Technology, Jamia Hamdard, New Delhi-110062 India
| |
Collapse
|
28
|
Innate Immune Response to Dengue Virus: Toll-like Receptors and Antiviral Response. Viruses 2022; 14:v14050992. [PMID: 35632732 PMCID: PMC9147118 DOI: 10.3390/v14050992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
Dengue is a mosquito-borne viral disease caused by the dengue virus (DENV1-4). The clinical manifestations range from asymptomatic to life-threatening dengue hemorrhagic fever (DHF) and/or Dengue Shock Syndrome (DSS). Viral and host factors are related to the clinical outcome of dengue, although the disease pathogenesis remains uncertain. The innate antiviral response to DENV is implemented by a variety of immune cells and inflammatory mediators. Blood monocytes, dendritic cells (DCs) and tissue macrophages are the main target cells of DENV infection. These cells recognize pathogen-associated molecular patterns (PAMPs) through pattern recognition receptors (PRRs). Pathogen recognition is a critical step in eliciting the innate immune response. Toll-like receptors (TLRs) are responsible for the innate recognition of pathogens and represent an essential component of the innate and adaptive immune response. Ten different TLRs are described in humans, which are expressed in many different immune cells. The engagement of TLRs with viral PAMPs triggers downstream signaling pathways leading to the production of inflammatory cytokines, interferons (IFNs) and other molecules essential for the prevention of viral replication. Here, we summarize the crucial TLRs’ roles in the antiviral innate immune response to DENV and their association with viral pathogenesis.
Collapse
|
29
|
Qiu L, Huang X, Luo J, Zhao Y, Hong S, Wang X, Feng K, Pan Y, Sun Q. Secondary cross infection with dengue virus serotype 2/3 aggravates vascular leakage in BALB/c mice. J Med Virol 2022; 94:4338-4347. [PMID: 35510565 DOI: 10.1002/jmv.27826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 11/09/2022]
Abstract
Dengue virus (DV) has occasionally emerged at epidemic levels in Yunnan, China. Vaccine development is limited by antibody dependent enhancement (ADE) and a lack of good animal models. Thus study investigated cross infection based on maternal immunity in BALB/c mice and assessed the risk of cross infection by DV2-D13113 and DV3-YNWS2 epidemic virus strains. DV replicated within the organs of the BALB/c infant mice, even causing death. Particularly, DV3-infected infant mice were at higher risk of severe disease if their mothers were infected with DV2. Although BALB/c adults and pups survived DV2/DV3 infection and produced anti-DV antibodies after 5-8 days, extensive subcutaneous vascular leakage was observed after secondary DV infection. Further, vascular permeability in the lung and kidney significantly increased in offspring born to heterotypic virus-infected mothers. Thus, vascular leakage indicates severe DV infection. The results indicate that maternal immunity increases the severity of subsequent heterotypic infection. Additionally, secondary cross infection by D13113 and YNWS2 represents a risk of serious disease. This study has implications for studies of DV cross infection and vaccine development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Lijuan Qiu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Kunming Children's Hospital, Yunnan Key Laboratory of Children's Major Disease Research, Yunnan Institute of Pediatrics, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Xinwei Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jia Luo
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Kunming Medical University Haiyuan College, Kunming, China
| | - Yujiao Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Shan Hong
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Xiaodan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Kai Feng
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China.,Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China.,Current postal address: Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMC), 935 Jiao Ling Road, Kunming, Yunnan, Province 650118, P.R, China
| |
Collapse
|
30
|
Generation and Characterization of Human-Mouse STING Chimeras That Allow DENV Replication in Mouse Cells. mSphere 2022; 7:e0091421. [PMID: 35477320 PMCID: PMC9241525 DOI: 10.1128/msphere.00914-21] [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: 11/20/2022] Open
Abstract
Our group was the first to describe direct antagonism of the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway by dengue virus (DENV) in human cells, and here, we report new findings on the characterization of the interaction between the DENV nonstructural protein 2B (NS2B)-NS3 (NS2B3) protease complex and STING. We demonstrate interactions between NS2B and the transmembrane domains of human STING and between NS3 and a portion of the cytoplasmic C-terminal domain of human STING. One significant obstacle we face today in the DENV field is the lack of small animal models available that can effectively recapitulate DENV pathogenesis in the early events of infection. The existing mouse models are either immunocompromised mice lacking interferon (IFN) receptors or "humanized" mice reconstituted with human stem cells. However, both approaches fail to capture important aspects of human pathogenesis because they lack critical innate immunity components or have deficiencies in immune cell development or maintenance. As an important step toward developing an immunocompetent mouse model for DENV, we have generated two chimeric human-mouse STING constructs that have promise in retaining both cleavability by NS2B3 and signaling capacity in the mouse. IMPORTANCE This article characterizes the interaction between human STING and DENV viral protease complex NS2B3 by constructing serial deletion mutants of STING. Our findings suggest that DENV nonstructural protein NS2B interacts with the transmembrane domains and NS3 with the C-terminal cyclic dinucleotide binding domain of human STING. Furthermore, as there exists no ideal immunocompetent murine model that can simultaneously support robust DENV replication and recapitulate the clinical manifestation of dengue disease observed in humans, we expressed and characterized two promising human-mouse chimeric STING constructs that can be used for developing a relevant transgenic mouse model to study dengue in the future. Both constructs can activate normal IFN responses in the overexpression system and be cleaved under infection conditions. We believe our findings offer a roadmap to the further development of a murine model that can greatly facilitate antiviral discoveries and vaccine research for DENV.
Collapse
|
31
|
Cantaert T, Jouvenet N, Diehl SA. Editorial: Balanced and Unbalanced Immune Response to Dengue Virus in Disease Protection and Pathogenesis. Front Immunol 2022; 13:835731. [PMID: 35222425 PMCID: PMC8873179 DOI: 10.3389/fimmu.2022.835731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- *Correspondence: Tineke Cantaert, ; Nolwenn Jouvenet, ; Sean A. Diehl,
| | - Nolwenn Jouvenet
- Institut Pasteur, Université de Paris, CNRS UMR3569, Virus Sensing and Signaling Unit, Paris, France
- *Correspondence: Tineke Cantaert, ; Nolwenn Jouvenet, ; Sean A. Diehl,
| | - Sean A. Diehl
- Department of Microbiology and Molecular Genetics, Translational Global Infectious Diseases Research Center, University of Vermont Larner College of Medicine, Burlington, VT, United States
- *Correspondence: Tineke Cantaert, ; Nolwenn Jouvenet, ; Sean A. Diehl,
| |
Collapse
|
32
|
Byrne AB, García CC, Damonte EB, Talarico LB. Murine models of dengue virus infection for novel drug discovery. Expert Opin Drug Discov 2022; 17:397-412. [PMID: 35098849 DOI: 10.1080/17460441.2022.2033205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Dengue virus (DENV) is the causative agent of the most prevalent human disease transmitted by mosquitoes in tropical and subtropical regions worldwide. At present, no antiviral drug is available and the difficulties to develop highly protective vaccines against the four DENV serotypes maintain the requirement of effective options for dengue chemotherapy. AREAS COVERED The availability of animal models that reproduce human disease is a very valuable tool for the preclinical evaluation of potential antivirals. Here, the main murine models of dengue infection are described, including immunocompetent wild-type mice, immunocompromised mice deficient in diverse components of the interferon (IFN) pathway and humanized mice. The main findings in antiviral testing of DENV inhibitory compounds in murine models are also presented. EXPERT OPINION At present, there is no murine model that fully recapitulates human disease. However, immunocompromised mice deficient in IFN-α/β and -γ receptors, with their limitations, have shown to be the most suitable system for antiviral preclinical testing. In fact, the AG129 mouse model allowed the identification of celgosivir, an inhibitor of cellular glucosidases, as a promising option for DENV therapy. However, clinical trials still were not successful, emphasizing the difficulties in the transition from preclinical testing to human treatment.
Collapse
Affiliation(s)
- Alana B Byrne
- Laboratorio de Investigaciones Infectológicas y Biología Molecular, Infectología, Departamento de Medicina, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cybele C García
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica-IQUIBICEN (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Elsa B Damonte
- Laboratorio de Estrategias Antivirales, Departamento de Química Biológica-IQUIBICEN (CONICET-UBA), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Laura B Talarico
- Laboratorio de Investigaciones Infectológicas y Biología Molecular, Infectología, Departamento de Medicina, Hospital de Niños Dr. Ricardo Gutiérrez, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
33
|
Ma J, Boudewijns R, Sanchez-Felipe L, Mishra N, Vercruysse T, Buh Kum D, Thibaut HJ, Neyts J, Dallmeier K. Comparing immunogenicity and protective efficacy of the yellow fever 17D vaccine in mice. Emerg Microbes Infect 2021; 10:2279-2290. [PMID: 34792431 PMCID: PMC8648041 DOI: 10.1080/22221751.2021.2008772] [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] [Indexed: 02/06/2023]
Abstract
The live-attenuated yellow fever 17D (YF17D) vaccine is one of the most efficacious human vaccines and also employed as a vector for novel vaccines. However, in the lack of appropriate immunocompetent small animal models, mechanistic insight in YF17D-induced protective immunity remains limited. To better understand YF17D vaccination and to identify a suitable mouse model, we evaluated the immunogenicity and protective efficacy of YF17D in five complementary mouse models, i.e. wild-type (WT) BALB/c, C57BL/6, IFN-α/β receptor (IFNAR-/-) deficient mice, and in WT mice in which type I IFN signalling was temporally ablated by an IFNAR blocking (MAR-1) antibody. Alike in IFNAR-/- mice, YF17D induced in either WT mice strong humoral immune responses dominated by IgG2a/c isotype (Th1 type) antibodies, yet only when IFNAR was blocked. Vigorous cellular immunity characterized by CD4+ T-cells producing IFN-γ and TNF-α were mounted in MAR-1 treated C57BL/6 and in IFNAR-/- mice. Surprisingly, vaccine-induced protection was largely mouse model dependent. Full protection against lethal intracranial challenge and a massive reduction of virus loads was conferred already by a minimal dose of 2 PFU YF17D in BALB/c and IFNAR-/- mice, but not in C57BL/6 mice. Correlation analysis of infection outcome with pre-challenge immunological markers indicates that YFV-specific IgG might suffice for protection, even in the absence of detectable levels of neutralizing antibodies. Finally, we propose that, in addition to IFNAR-/- mice, C57BL/6 mice with temporally blocked IFN-α/β receptors represent a promising immunocompetent mouse model for the study of YF17D-induced immunity and evaluation of YF17D-derived vaccines.
Collapse
Affiliation(s)
- Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Robbert Boudewijns
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Niraj Mishra
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Thomas Vercruysse
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Dieudonné Buh Kum
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium
| | - Hendrik Jan Thibaut
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Rega Institute, Leuven, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,Global Virus Network (GVN), Baltimore, MD, USA
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Rega Institute, Leuven, Belgium.,Global Virus Network (GVN), Baltimore, MD, USA
| |
Collapse
|
34
|
Chen RE, Smith BK, Errico JM, Gordon DN, Winkler ES, VanBlargan LA, Desai C, Handley SA, Dowd KA, Amaro-Carambot E, Cardosa MJ, Sariol CA, Kallas EG, Sékaly RP, Vasilakis N, Fremont DH, Whitehead SS, Pierson TC, Diamond MS. Implications of a highly divergent dengue virus strain for cross-neutralization, protection, and vaccine immunity. Cell Host Microbe 2021; 29:1634-1648.e5. [PMID: 34610295 PMCID: PMC8595868 DOI: 10.1016/j.chom.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/15/2021] [Accepted: 09/10/2021] [Indexed: 01/29/2023]
Abstract
Although divergent dengue viruses (DENVs) have been isolated in insects, nonhuman primates, and humans, their relationships to the four canonical serotypes (DENV 1-4) are poorly understood. One virus isolated from a dengue patient, DKE-121, falls between genotype and serotype levels of sequence divergence to DENV-4. To examine its antigenic relationship to DENV-4, we assessed serum neutralizing and protective activity. Whereas DENV-4-immune mouse sera neutralize DKE-121 infection, DKE-121-immune sera inhibit DENV-4 less efficiently. Passive transfer of DENV-4 or DKE-121-immune sera protects mice against homologous, but not heterologous, DENV-4 or DKE-121 challenge. Antigenic cartography suggests that DENV-4 and DKE-121 are related but antigenically distinct. However, DENV-4 vaccination confers protection against DKE-121 in nonhuman primates, and serum from humans immunized with a tetravalent vaccine neutralize DENV-4 and DKE-121 infection equivalently. As divergent DENV strains, such as DKE-121, may meet criteria for serotype distinction, monitoring their capacity to impact dengue disease and vaccine efficacy appears warranted.
Collapse
Affiliation(s)
- Rita E Chen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Brittany K Smith
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - John M Errico
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - David N Gordon
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-9806, USA
| | - Emma S Winkler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Laura A VanBlargan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Chandni Desai
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Scott A Handley
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Kimberly A Dowd
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-9806, USA
| | - Emerito Amaro-Carambot
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-9806, USA
| | - M Jane Cardosa
- Institute of Health and Community Medicine, Universiti Sarawak Malaysia (UNIMAS), Kota Samarahan, Sarawak 94300, Malaysia; Integrated Research Associates, San Rafael, CA 94903, USA
| | - Carlos A Sariol
- Unit of Comparative Medicine, Caribbean Primate Research Center, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00936-5067, USA
| | - Esper G Kallas
- Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Rafick-Pierre Sékaly
- Department of Microbiology and Immunology, Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nikos Vasilakis
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Vector-Borne and Zoonotic Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; The Andrew M. Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110-1010, USA
| | - Stephen S Whitehead
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-9806, USA
| | - Theodore C Pierson
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-9806, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; The Andrew M. Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63110-1010, USA; Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, Saint Louis, MO 63110-1010, USA.
| |
Collapse
|
35
|
de Mendonça SF, Rocha MN, Ferreira FV, Leite THJF, Amadou SCG, Sucupira PHF, Marques JT, Ferreira AGA, Moreira LA. Evaluation of Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus Mosquitoes Competence to Oropouche virus Infection. Viruses 2021; 13:v13050755. [PMID: 33923055 PMCID: PMC8145018 DOI: 10.3390/v13050755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/22/2022] Open
Abstract
The emergence of new human viral pathogens and re-emergence of several diseases are of particular concern in the last decades. Oropouche orthobunyavirus (OROV) is an arbovirus endemic to South and Central America tropical regions, responsible to several epidemic events in the last decades. There is little information regarding the ability of OROV to be transmitted by urban/peri-urban mosquitoes, which has limited the predictability of the emergence of permanent urban transmission cycles. Here, we evaluated the ability of OROV to infect, replicate, and be transmitted by three anthropophilic and urban species of mosquitoes, Aedes aegypti, Aedes albopictus, and Culex quinquefasciatus. We show that OROV is able to infect and efficiently replicate when systemically injected in all three species tested, but not when orally ingested. Moreover, we find that, once OROV replication has occurred in the mosquito body, all three species were able to transmit the virus to immunocompromised mice during blood feeding. These data provide evidence that OROV is restricted by the midgut barrier of three major urban mosquito species, but, if this restriction is overcome, could be efficiently transmitted to vertebrate hosts. This poses a great risk for the emergence of permanent urban cycles and geographic expansion of OROV to other continents.
Collapse
Affiliation(s)
- Silvana F. de Mendonça
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - Marcele N. Rocha
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Flávia V. Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Thiago H. J. F Leite
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Siad C. G. Amadou
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
| | - Pedro H. F. Sucupira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - João T. Marques
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, 6627-Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.V.F.); (T.H.J.F.L.); (S.C.G.A.); (J.T.M.)
- Faculté des Sciences de laVie, Université de Strasbourg, CNRS UPR9022, Inserm U1257, 67084 Strasbourg, France
| | - Alvaro G. A. Ferreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
| | - Luciano A. Moreira
- Mosquitos Vetores: Endossimbiontes e Interação Patógeno-Vetor, Instituto René Rachou—Fiocruz, Belo Horizonte 30190-002, MG, Brazil; (S.F.d.M.); (M.N.R.); (P.H.F.S.); (A.G.A.F.)
- Correspondence:
| |
Collapse
|
36
|
Pathogen Dose in Animal Models of Hemorrhagic Fever Virus Infections and the Potential Impact on Studies of the Immune Response. Pathogens 2021; 10:pathogens10030275. [PMID: 33804381 PMCID: PMC7999429 DOI: 10.3390/pathogens10030275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Viral hemorrhagic fever viruses come from a wide range of virus families and are a significant cause of morbidity and mortality worldwide each year. Animal models of infection with a number of these viruses have contributed to our knowledge of their pathogenesis and have been crucial for the development of therapeutics and vaccines that have been approved for human use. Most of these models use artificially high doses of virus, ensuring lethality in pre-clinical drug development studies. However, this can have a significant effect on the immune response generated. Here I discuss how the dose of antigen or pathogen is a critical determinant of immune responses and suggest that the current study of viruses in animal models should take this into account when developing and studying animal models of disease. This can have implications for determination of immune correlates of protection against disease as well as informing relevant vaccination and therapeutic strategies.
Collapse
|
37
|
A single-dose live attenuated chimeric vaccine candidate against Zika virus. NPJ Vaccines 2021; 6:20. [PMID: 33514743 PMCID: PMC7846741 DOI: 10.1038/s41541-021-00282-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
The mosquito-borne Zika virus is an emerging pathogen from the Flavivirus genus for which there are no approved antivirals or vaccines. Using the clinically validated PDK-53 dengue virus vaccine strain as a backbone, we created a chimeric dengue/Zika virus, VacDZ, as a live attenuated vaccine candidate against Zika virus. VacDZ demonstrates key markers of attenuation: small plaque phenotype, temperature sensitivity, attenuation of neurovirulence in suckling mice, and attenuation of pathogenicity in interferon deficient adult AG129 mice. VacDZ may be administered as a traditional live virus vaccine, or as a DNA-launched vaccine that produces live VacDZ in vivo after delivery. Both vaccine formulations induce a protective immune response against Zika virus in AG129 mice, which includes neutralising antibodies and a strong Th1 response. This study demonstrates that VacDZ is a safe and effective vaccine candidate against Zika virus.
Collapse
|
38
|
Coelho SVA, Rust NM, Vellasco L, Papa MP, Pereira ASG, da Silva Palazzo MF, Juliano MA, Costa SM, Alves AMB, Cordeiro MT, Marques ETA, Scharfstein J, de Arruda LB. Contact System Activation in Plasma from Dengue Patients Might Harness Endothelial Virus Replication through the Signaling of Bradykinin Receptors. Pharmaceuticals (Basel) 2021; 14:ph14010056. [PMID: 33445640 PMCID: PMC7827195 DOI: 10.3390/ph14010056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023] Open
Abstract
Since exacerbated inflammation and microvascular leakage are hallmarks of dengue virus (DENV) infection, here we interrogated whether systemic activation of the contact/kallikrein-kinin system (KKS) might hamper endothelial function. In vitro assays showed that dextran sulfate, a potent contact activator, failed to generate appreciable levels of activated plasma kallikrein (PKa) in the large majority of samples from a dengue cohort (n = 70), irrespective of severity of clinical symptoms. Impaired formation of PKa in dengue-plasmas correlated with the presence of cleaved Factor XII and high molecular weight kininogen (HK), suggesting that the prothrombogenic contact system is frequently triggered during the course of infection. Using two pathogenic arboviruses, DENV or Zika virus (ZIKV), we then asked whether exogenous BK could influence the outcome of infection of human brain microvascular endothelial cells (HBMECs). Unlike the unresponsive phenotype of Zika-infected HBMECs, we found that BK, acting via B2R, vigorously stimulated DENV-2 replication by reverting nitric oxide-driven apoptosis of endothelial cells. Using the mouse model of cerebral dengue infection, we next demonstrated that B2R targeting by icatibant decreased viral load in brain tissues. In summary, our study suggests that contact/KKS activation followed by BK-induced enhancement of DENV replication in the endothelium may underlie microvascular pathology in dengue.
Collapse
Affiliation(s)
- Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Naiara M. Rust
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Lucas Vellasco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Aline S. G. Pereira
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
| | - Matheus Ferreira da Silva Palazzo
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
| | - Maria Aparecida Juliano
- Departamento de Biofísica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil;
| | - Simone M. Costa
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Ada M. B. Alves
- Laboratório de Biotecnologia e Fisiologia de Infecções Virais, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, Brazil; (S.M.C.); (A.M.B.A.)
| | - Marli T. Cordeiro
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
| | - Ernesto T. A. Marques
- Fundação Oswaldo Cruz, Instituto Aggeu Magalhães, Recife 50740-465, Brazil; (M.T.C.); (E.T.A.M.)
- Department of Infectious Diseases, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Júlio Scharfstein
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (M.F.d.S.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (S.V.A.C.); (N.M.R.); (M.P.P.); (A.S.G.P.)
- Correspondence: (J.S.); (L.B.d.A.)
| |
Collapse
|