1
|
Muccilli SG, Schwarz B, Shue B, Jessop F, Shannon JG, Larson CL, Hage A, Hong SH, Bohrnsen E, Hsu T, Ashbrook AW, Sturdevant GL, Robertson SJ, Guarnieri JW, Lack J, Wallace DC, Bosio CM, MacDonald MR, Rice CM, Yewdell JW, Best SM. Mitochondrial hyperactivity and reactive oxygen species drive innate immunity to the yellow fever virus-17D live-attenuated vaccine. PLoS Pathog 2025; 21:e1012561. [PMID: 40258014 PMCID: PMC12052391 DOI: 10.1371/journal.ppat.1012561] [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: 09/03/2024] [Revised: 05/05/2025] [Accepted: 03/31/2025] [Indexed: 04/23/2025] Open
Abstract
The yellow fever virus 17D (YFV-17D) live attenuated vaccine is considered one of the most successful vaccines ever generated associated with high antiviral immunity, yet the signaling mechanisms that drive the response in infected cells are not understood. Here, we provide a molecular understanding of how metabolic stress and innate immune responses are linked to drive type I IFN expression in response to YFV-17D infection. Comparison of YFV-17D replication with its parental virus, YFV-Asibi, and a related dengue virus revealed that IFN expression requires RIG-I-Like Receptor signaling through MAVS, as expected. However, YFV-17D uniquely induces mitochondrial respiration and major metabolic perturbations, including hyperactivation of electron transport to fuel ATP synthase. Mitochondrial hyperactivity generates reactive oxygen species (ROS) including peroxynitrite, blocking of which abrogated MAVS oligomerization and IFN expression in non-immune cells without reducing YFV-17D replication. Scavenging ROS in YFV-17D-infected human dendritic cells increased cell viability yet globally prevented expression of IFN signaling pathways. Thus, adaptation of YFV-17D for high growth imparts mitochondrial hyperactivity to meet energy demands, resulting in generation of ROS as the critical messengers that convert a blunted IFN response into maximal activation of innate immunity essential for vaccine effectiveness.
Collapse
Affiliation(s)
- Samantha G. Muccilli
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Benjamin Schwarz
- Research Technologies Branch, NIAID, NIH, Hamilton, Montana, United States of America
| | - Byron Shue
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Forrest Jessop
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, Montana, United States of America
| | - Jeffrey G. Shannon
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Charles L. Larson
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Adam Hage
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Seon-Hui Hong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Eric Bohrnsen
- Research Technologies Branch, NIAID, NIH, Hamilton, Montana, United States of America
| | - Thomas Hsu
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Alison W. Ashbrook
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Gail L. Sturdevant
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Shelly J. Robertson
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Division on Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, Montana, United States of America
| | - Margaret R. MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Jonathan W. Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Sonja M. Best
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| |
Collapse
|
2
|
Zaib S, Rana N, Ali HS, Ur Rehman M, Awwad NS, Ibrahium HA, Khan I. Identification of potential inhibitors targeting yellow fever virus helicase through ligand and structure-based computational studies. J Biomol Struct Dyn 2025; 43:3031-3048. [PMID: 38109183 DOI: 10.1080/07391102.2023.2294839] [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: 08/14/2023] [Accepted: 12/05/2023] [Indexed: 12/19/2023]
Abstract
Yellow fever is a flavivirus having plus-sensed RNA which encodes a single polyprotein. Host proteases cut this polyprotein into seven nonstructural proteins including a vital NS3 protein. The present study aims to identify the most effective inhibitor against the helicase (NS3) using different advanced ligand and structure-based computational studies. A set of 300 ligands was selected against helicase by chemical structural similarity model, which are similar to S-adenosyl-l-cysteine using infiniSee. This tool screens billions of compounds through a similarity search from in-built chemical spaces (CHEMriya, Galaxi, KnowledgeSpace and REALSpace). The pharmacophore was designed from ligands in the library that showed same features. According to the sequence of ligands, six compounds (29, 87, 99, 116, 148, and 208) were taken for pharmacophore designing against helicase protein. Subsequently, compounds from the library which showed the best pharmacophore shared-features were docked using FlexX functionality of SeeSAR and their optibrium properties were analyzed. Afterward, their ADME was improved by replacing the unfavorable fragments, which resulted in the generation of new compounds. The selected best compounds (301, 302, 303 and 304) were docked using SeeSAR and their pharmacokinetics and toxicological properties were evaluated using SwissADME. The optimal inhibitor for yellow fever helicase was 2-amino-N-(4-(dimethylamino)thiazol-2-yl)-4-methyloxazole-5-carboxamide (302), which exhibits promising potential for drug development.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Nehal Rana
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Hafiz Saqib Ali
- Chemistry Research Laboratory, Department of Chemistry and the INEOS Oxford Institute for Antimicrobial Research, University of Oxford, Oxford, UK
| | - Mujeeb Ur Rehman
- Department of Basic and Applied Chemistry, Faculty of Science and Technology, University of Central Punjab, Lahore, Pakistan
| | - Nasser S Awwad
- Department of Chemistry, King Khalid University, Abha, Saudi Arabia
| | - Hala A Ibrahium
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Imtiaz Khan
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, UK
| |
Collapse
|
3
|
Cenci Dietrich V, Costa JMC, Oliveira MMGL, Aguiar CEO, Silva LGDO, Luz MS, Lemos FFB, de Melo FF. Pathogenesis and clinical management of arboviral diseases. World J Virol 2025; 14:100489. [PMID: 40134841 PMCID: PMC11612872 DOI: 10.5501/wjv.v14.i1.100489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Arboviral diseases are viral infections transmitted to humans through the bites of arthropods, such as mosquitoes, often causing a variety of pathologies associated with high levels of morbidity and mortality. Over the past decades, these infections have proven to be a significant challenge to health systems worldwide, particularly following the considerable geographic expansion of the dengue virus (DENV) and its most recent outbreak in Latin America as well as the difficult-to-control outbreaks of yellow fever virus (YFV), chikungunya virus (CHIKV), and Zika virus (ZIKV), leaving behind a substantial portion of the population with complications related to these infections. Currently, the world is experiencing a period of intense globalization, which, combined with global warming, directly contributes to wider dissemination of arbovirus vectors across the globe. Consequently, all continents remain on high alert for potential new outbreaks. Thus, this review aims to provide a comprehensive understanding of the pathogenesis of the four main arboviruses today (DENV, ZIKV, YFV, and CHIKV) discussing their viral characteristics, immune responses, and mechanisms of viral evasion, as well as important clinical aspects for patient management. This includes associated symptoms, laboratory tests, treatments, existing or developing vaccines and the main associated complications, thus integrating a broad historical, scientific and clinical approach.
Collapse
Affiliation(s)
- Victoria Cenci Dietrich
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Juan Marcos Caram Costa
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | | | | | - Marcel Silva Luz
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabian Fellipe Bueno Lemos
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Instituto Multidisciplinar em Saúde, Universidade Federal da Bahia, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
4
|
Handa T, Saha A, Narayanan A, Ronzier E, Kumar P, Singla J, Tomar S. Structural Virology: The Key Determinants in Development of Antiviral Therapeutics. Viruses 2025; 17:417. [PMID: 40143346 PMCID: PMC11945554 DOI: 10.3390/v17030417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Structural virology has emerged as the foundation for the development of effective antiviral therapeutics. It is pivotal in providing crucial insights into the three-dimensional frame of viruses and viral proteins at atomic-level or near-atomic-level resolution. Structure-based assessment of viral components, including capsids, envelope proteins, replication machinery, and host interaction interfaces, is instrumental in unraveling the multiplex mechanisms of viral infection, replication, and pathogenesis. The structural elucidation of viral enzymes, including proteases, polymerases, and integrases, has been essential in combating viruses like HIV-1 and HIV-2, SARS-CoV-2, and influenza. Techniques including X-ray crystallography, Nuclear Magnetic Resonance spectroscopy, Cryo-electron Microscopy, and Cryo-electron Tomography have revolutionized the field of virology and significantly aided in the discovery of antiviral therapeutics. The ubiquity of chronic viral infections, along with the emergence and reemergence of new viral threats necessitate the development of novel antiviral strategies and agents, while the extensive structural diversity of viruses and their high mutation rates further underscore the critical need for structural analysis of viral proteins to aid antiviral development. This review highlights the significance of structure-based investigations for bridging the gap between structure and function, thus facilitating the development of effective antiviral therapeutics, vaccines, and antibodies for tackling emerging viral threats.
Collapse
Affiliation(s)
- Tanuj Handa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Ankita Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Aarthi Narayanan
- Department of Biology, College of Science, George Mason University, Fairfax, VA 22030, USA;
| | - Elsa Ronzier
- Biomedical Research Laboratory, Institute for Biohealth Innovation, George Mason University, Fairfax, VA 22030, USA;
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Jitin Singla
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India; (T.H.); (A.S.); (P.K.); (J.S.)
| |
Collapse
|
5
|
Le Breton C, Laporta GZ, Sallum MAM, Hesse H, Salgado-Lynn M, Manin BO, Fornace K. Advancing canopy-level entomological surveillance to monitor vector-borne and zoonotic disease dynamics. Trends Parasitol 2025; 41:150-161. [PMID: 39809620 DOI: 10.1016/j.pt.2024.12.009] [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: 10/26/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Faced with the increased frequency of zoonotic spillover in recent decades, emerging vector-borne diseases from nonhuman primates pose a significant threat to global public health. Understanding transmission dynamics driven by arthropod vectors between wildlife populations is critical for surveillance, modeling, and mitigation. Elevated canopy-level sampling is a valuable approach for elucidating vector behavior and sylvatic transmission. However, this is underused in many regions because of the logistical and mechanical challenges of repurposing ground-based trapping for the forest canopy. We review methods of canopy-level entomological surveillance, present case studies, and identify opportunities to integrate new technologies. Paired with robust experimental design, canopy-level trapping can complement existing surveillance of emerging zoonotic diseases and provide critical insights into the role of vectors driving spillover risks.
Collapse
Affiliation(s)
| | - Gabriel Z Laporta
- Graduate Research and Innovation Program, Centro Universitario FMABC, Santo André, São Paulo, Brazil
| | - Maria Anice Mureb Sallum
- Departamento de Epidemiologia, Faculdade de Saude Publica, Universidade de São Paulo, São Paulo, Brazil
| | - Henrik Hesse
- James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Milena Salgado-Lynn
- Danau Girang Field Centre, c/o Sabah Wildlife Department, Sabah, Malaysia; School of Biosciences, Cardiff University, Cardiff, UK
| | | | - Kimberly Fornace
- Saw Swee Hock School of Public Health and National University Health System, National University of Singapore, Singapore.
| |
Collapse
|
6
|
Pastor F, Delphin M, Lucifora J, Verrier ER. [Non-alphabetic viral hepatitis]. Med Sci (Paris) 2025; 41:145-153. [PMID: 40028952 DOI: 10.1051/medsci/2025010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
The liver is the target of various viruses that can cause significant damage, impair function and potentially threaten a patient's life. While the "alphabetic" hepatitis viruses A, B, C, D, and E are well-characterized, and their impact on liver function well-documented, many emerging and re-emerging viruses, some of which are considered by the WHO to be potential pandemic threats, also infect the liver. In this review, we describe the current state of knowledge regarding liver infections caused by major non-alphabetic hepatotropic viruses and their effects on liver functions.
Collapse
Affiliation(s)
- Florentin Pastor
- CIRI, Centre international de recherche en infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | | | - Julie Lucifora
- CIRI, Centre international de recherche en infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Eloi R Verrier
- Université de Strasbourg, Inserm, ITM UMR_S1110, Strasbourg, France
| |
Collapse
|
7
|
Garcia HLP. Epidemic Outbreaks Related to Yellow Fever Viruses. Methods Mol Biol 2025; 2913:251-266. [PMID: 40249443 DOI: 10.1007/978-1-0716-4458-4_24] [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] [Indexed: 04/19/2025]
Abstract
Epidemic outbreaks related to yellow fever viruses, added to the climate of fear resulting from the COVID-19 pandemic, raise a spectrum of concern and need for preparation in different research, epidemiology, and health groups. This question refers to conditions of: molecular analysis of the virus, its dissemination potential, vectors, mutational speed rate, evolutionary potential, and potential damage to individuals and populations.With regard to the yellow fever virus, such questions are even more pressing due to its successful history of colonization in America, originating from regions of Africa, and becoming endemic in both continents.Through the study of viral history and epidemiology, we seek to establish bases that allow us to analyze or avoid future problems and questions.This analysis of the past generates a future fear associated with a very real possibility:What are the chances of yellow fever spreading in densely populated regions of Asia?What are the chances of the emergence of a new pandemic caused by a virus that is already so well established on the planet?And in an associated way, the questions arise: How to avoid the possibility of a new epidemic? Is the vaccine associated with defense against this virus effective? Is the production of such a vaccine reliable and widespread in case of future deleterious events? All of these issues are relevant and worthy of analysis and response in future events, with a view to maximizing the health of the general population and mitigating human and economic damage.
Collapse
|
8
|
Tang Y, Li Y, Cai X, Yin X. Viral Live-Attenuated Vaccines (LAVs): Past and Future Directions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407241. [PMID: 39639853 PMCID: PMC11744563 DOI: 10.1002/advs.202407241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Viral infections continue to pose a significant threat to the health of both humans and animals. Currently, live attenuated vaccines (LAVs) remain the most efficacious and widely utilized tool for combating viral infections. Conventional LAVs involve the adaptation of virulent viruses to novel hosts, cell cultures, or suboptimal environments, resulting in a reduction in pathogenicity while retaining immunogenicity. This process entails directed evolution of the virus to enhance its replication efficiency under these modified conditions. In this review, the development of traditional animal-adapted and cold-adapted LAVs is specially discussed. Additionally, the factors that contribute to virus attenuation from a viral lifecycle perspective are summarized. Finally, we propose future directions for next-generation LAVs.
Collapse
Affiliation(s)
- Yan‐Dong Tang
- State Key Laboratory for Animal Disease Control and PreventionHarbin Veterinary Research Institute of Chinese Academy of Agricultural SciencesHarbin150069China
- Heilongjiang Provincial Research Center for Veterinary BiomedicineHarbin Veterinary Research Institute of Chinese Academy of Agricultural SciencesHarbin150069China
- Heilongjiang Provincial Key Laboratory of Veterinary ImmunologyHarbin Veterinary Research Institute of Chinese Academy of Agricultural SciencesHarbin150069China
| | - Yuming Li
- School of Public HealthShandong First Medical University & Shandong Academy of Medical SciencesJi'nan250117China
- Key Laboratory of Emerging Infectious Diseases in Universities of ShandongShandong First Medical University & Shandong Academy of Medical SciencesTai'an271000China
| | - Xue‐Hui Cai
- State Key Laboratory for Animal Disease Control and PreventionHarbin Veterinary Research Institute of Chinese Academy of Agricultural SciencesHarbin150069China
| | - Xin Yin
- State Key Laboratory for Animal Disease Control and PreventionHarbin Veterinary Research Institute of Chinese Academy of Agricultural SciencesHarbin150069China
| |
Collapse
|
9
|
de Lima RC, da Costa Faria NR, de Carvalho AT. Flow Cytometry as Immunoassay Tool for Research on Yellow Fever Virus. Methods Mol Biol 2025; 2913:1-17. [PMID: 40249420 DOI: 10.1007/978-1-0716-4458-4_1] [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] [Indexed: 04/19/2025]
Abstract
Flow cytometry is a sensitive and practical technique that can be applied in both basic and clinical research. It allows extracting quantitative and multiparametric valuable information to assist in the study of cell immunophenotyping, morphological complexity, location and/or expression of extra and intracellular molecules involved in metabolic and proliferative pathways, inflammation, viability, and cell death, among others. The parameters are analyzed by labeling antigens of yellow fever virus and/or cells with fluorescent specific monoclonal antibodies or dyes for immunophenotyping and data acquisition using a flow cytometer. In translational research, flow cytometry is a useful tool in tracking and monitoring the etiology, evolution, and outcome of several infectious diseases, such as yellow fever (YF), with the aim of benefiting human health through detection of intracellular viral antigen and vaccine efficacy trials, as well as characterization, quantification, and monitoring of immune cells subpopulations and their biological functions quality. In the last 10 years we have been facing the re-emergence of YF, considered an endemic disease caused by arbovirus in continents including South America. Unfortunately, severe forms of the disease are still associated with increased mortality. Even with the availability of effective vaccines, gaps about understanding the immune pathophysiology and clinical management are still considered a huge challenge for the scientific community. Therefore, such tool has the potential to aggregate in flavivirus setting, being effective in tracking infection in different biological culture systems, animal and human models, as well as in searching for new antiviral drugs and vaccine efficacy.
Collapse
Affiliation(s)
- Raquel Curtinhas de Lima
- Laboratory of Parasite-Host Interactions (LIVH), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Nieli Rodrigues da Costa Faria
- Laboratory of Arboviruses and Hemorrhagic Viruses (LARBOH), Oswaldo Cruz Institute (IOC), Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Amanda Torrentes de Carvalho
- Biology Institute, Department of Imunobiology (GIM) Laboratory of Immunobiology of Infectious Diseases (LIDI), Fluminense Federal University (UFF), Niterói, Brazil
| |
Collapse
|
10
|
dos Santos CR, dos Santos CGM, Couto-Lima D, Souza BS, Rahman RU, Dornelas Ribeiro M, Lima JBP, Martins AJ. Evaluation of Yellow Fever Virus Infection in Aedes aegypti Mosquitoes from Pakistan with Distinct Knockdown Resistance Genotypes. INSECTS 2024; 16:33. [PMID: 39859614 PMCID: PMC11765701 DOI: 10.3390/insects16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Yellow fever (YF) is an acute hemorrhagic disease endemic to Africa and Latin America; however, no cases have been reported in Asian regions with high Aedes aegypti infestation. Factors such as environmental conditions and genetic variations in the yellow fever virus (YFV) strains and mosquito populations may explain this absence. Mosquito populations have undergone strong selective pressure owing to the excessive use of insecticides. This pressure has led to the spread of alterations, such as knockdown-resistant mutations (kdr), which, while conferring resistance to pyrethroids, also induce various physiological side effects in the insect. Therefore, it is important to investigate whether the presence of kdr mutations influences the infectivity of YFV mosquitoes. This study evaluated the susceptibility of Ae. aegypti from Pakistan with distinct kdr genotypes to different YFV strains under laboratory conditions. METHODS Ae. aegypti from a Pakistani colony were exposed to YFV strains (PR4408/2008 and ES504/2017) along with the Rockefeller strain. After 14 days, RNA and DNA were extracted for viral RNA detection (qPCR) and kdr genotyping (TaqMan qPCR and HRM for T1520I and F1534C SNPs). RESULTS Pakistani Ae. aegypti were orally susceptible to YFV, with infection rates of 83.7% (PR4408/2008) and 61.3% (ES504), respectively, similar to Rockefeller. Two kdr genotypes (II + CC and TI + FC) were identified, with no significant differences in viral infection or dissemination rates. CONCLUSIONS The Ae. aegypti population from Asia is capable of YFV infection and dissemination, regardless of kdr genotype.
Collapse
Affiliation(s)
- Carlucio Rocha dos Santos
- Laboratório de Biologia, Controlee Vigilância de InsetosVetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil (B.S.S.); (R.U.R.)
| | | | - Dinair Couto-Lima
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil
| | - Bárbara Silva Souza
- Laboratório de Biologia, Controlee Vigilância de InsetosVetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil (B.S.S.); (R.U.R.)
| | - Rafi Ur Rahman
- Laboratório de Biologia, Controlee Vigilância de InsetosVetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil (B.S.S.); (R.U.R.)
| | - Marcos Dornelas Ribeiro
- Instituto de Biologia do Exército, Divisão de Ensino e Pesquisa, Rio de Janeiro 20911-270, RJ, Brazil (M.D.R.)
| | - José Bento Pereira Lima
- Laboratório de Biologia, Controlee Vigilância de InsetosVetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil (B.S.S.); (R.U.R.)
| | - Ademir Jesus Martins
- Laboratório de Biologia, Controlee Vigilância de InsetosVetores, Instituto Oswaldo Cruz, FIOCRUZ, Rio de Janeiro 21040-360, RJ, Brazil (B.S.S.); (R.U.R.)
- Instituto Nacional de Ciência e Tecnologia (INCT), Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| |
Collapse
|
11
|
Libardi Lira Machado KL, da Costa-Rocha IA, Gonçalves Rodrigues Aguiar L, Ribeiro Moulaz I, Tatiyama Miyamoto S, Costa Martins P, Vieira Serrano E, Espíndula Gianordoli AP, da Penha Gomes Gouvea M, de Fatima Bissoli M, Maria Barbosa de Lima S, Dias Schwarcz W, de Souza Azevedo A, Fernandes Amorim da Silva J, Tourinho Santos R, Pedro Brito-de-Sousa J, Coelho-dos-Reis JG, Campi-Azevedo AC, Teixeira-Carvalho A, Peruhype-Magalhães V, Fontana Sutile Tardetti Fantinato F, Maria Henrique da Mota L, Assis Martins-Filho O, Valim V. Hydroxychloroquine is associated with lower seroconversion upon 17DD-Yellow fever primovaccination in patients with primary Sjögren's syndrome. Hum Vaccin Immunother 2024; 20:2318814. [PMID: 38961639 PMCID: PMC11225917 DOI: 10.1080/21645515.2024.2318814] [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: 11/20/2023] [Accepted: 02/10/2024] [Indexed: 07/05/2024] Open
Abstract
The present study aimed at investigating whether the hydroxychloroquine (HCQ) treatment would impact the neutralizing antibody production, viremia levels and the kinetics of serum soluble mediators upon planned 17DD-Yellow Fever (YF) primovaccination (Bio-Manguinhos-FIOCRUZ) of primary Sjögren's syndrome (pSS). A total of 34 pSS patients and 23 healthy controls (HC) were enrolled. The pSS group was further categorized according to the use of HCQ (HCQ and Non-HCQ). The YF-plaque reduction neutralization test (PRNT ≥1:50), YF viremia (RNAnemia) and serum biomarkers analyses were performed at baseline and subsequent time-points (Day0/Day3-4/Day5-6/Day7/Day14-D28). The pSS group showed PRNT titers and seropositivity rates similar to those observed for HC (GeoMean = 238 vs 440, p = .11; 82% vs 96%, p = .13). However, the HCQ subgroup exhibited lower seroconversion rates as compared to HC (GeoMean = 161 vs 440, p = .04; 69% vs 96%, p = .02) and Non-HQC (GeoMean = 161 vs 337, p = .582; 69% vs 94%, p = .049). No differences in YF viremia were observed amongst subgroups. Serum biomarkers analyses demonstrated that HCQ subgroup exhibited increased levels of CCL2, CXL10, IL-6, IFN-γ, IL1-Ra, IL-9, IL-10, and IL-2 at baseline and displayed a consistent increase of several biomarkers along the kinetics timeline up to D14-28. These results indicated that HCQ subgroup exhibited a deficiency in assembling YF-specific immune response elicited by 17DD-YF primovaccination as compared to Non-HCQ subgroup. Our findings suggested that hydroxychloroquine is associated with a decrease in the humoral immune response after 17DD-YF primovaccination.
Collapse
Affiliation(s)
- Ketty Lysie Libardi Lira Machado
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Ismael Artur da Costa-Rocha
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | - Laura Gonçalves Rodrigues Aguiar
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Isac Ribeiro Moulaz
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Samira Tatiyama Miyamoto
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Priscila Costa Martins
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Erica Vieira Serrano
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Ana Paula Espíndula Gianordoli
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Maria da Penha Gomes Gouvea
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Maria de Fatima Bissoli
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| | - Sheila Maria Barbosa de Lima
- Departamento de Desenvolvimento Experimental e Pré-clínico (DEDEP), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Waleska Dias Schwarcz
- Laboratório de Análise Imunomolecular (LANIM), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Adriana de Souza Azevedo
- Laboratório de Análise Imunomolecular (LANIM), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Juliana Fernandes Amorim da Silva
- Laboratório de Análise Imunomolecular (LANIM), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Renata Tourinho Santos
- Laboratório de Tecnologia Virológica (LATEV), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Joaquim Pedro Brito-de-Sousa
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | - Jordana Grazziela Coelho-dos-Reis
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Carolina Campi-Azevedo
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | | | - Licia Maria Henrique da Mota
- Serviço de Reumatologia do Hospital Universitário de Brasília, Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Brazil
| | - Valéria Valim
- Programa de Pós-Graduação em Saúde Coletiva (PPGSC) and Hospital Universitário Cassiano Antônio Moraes da Universidade Federal do Espírito Santo (HUCAM-UFES/EBSERH)
| |
Collapse
|
12
|
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
|
13
|
Van Herreweghe M, De Bruyne T, Hermans N, Huits R. Clinical Relevance of Oxidative Stress Biomarkers in Human Flavivirus Infections as Predictors of Disease Progression and Severity. Rev Med Virol 2024; 34:e70007. [PMID: 39532693 DOI: 10.1002/rmv.70007] [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: 04/08/2024] [Revised: 09/18/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Several Flaviviridae constitute an emerging threat to global health because of their continuing spread and the expansion of vector habitats, largely driven by climate change and intensified global travel. Infections can result in severe neurological or visceral pathologies. The relationship between oxidative stress (OS), an imbalance between generated reactive oxygen species and the antioxidant defences of the host, and flavivirus infection has been repeatedly demonstrated in in vitro and animal studies, but measuring biomarkers of oxidative stress in vivo could prove useful in clinical patient management. We summarise the knowledge and prospects of measuring peripheral OS biomarker levels for clinical case management and correlation with disease severity in six important human flavivirus infections (dengue virus (DENV), Japanese encephalitis virus, West Nile virus (WNV), tick-borne encephalitis virus (TBEV), yellow fever virus and zika virus). We searched the Medline and Web of Science databases for 'Oxidative Stress' AND 'Biomarkers' AND 'Flavivirus', combined with 'clinical', 'in vivo/in vivo', 'patient' and/or 'disease' and included 43 peer-reviewed publications. Correlation between OS and infection has been studied in all six Flaviviridae, but most clinically relevant data are available for DENV, TBEV and WNV. Plasma protein carbonyls, glutathione peroxidase activity and nitrogen monoxide are promising prognostic markers, but their measurement would benefit from methodological harmonisation. Future studies should investigate a broad range of OS biomarkers as predictors of clinically relevant outcomes. We advocate the validation and use of universal or disease-specific oxidative stress indexes that incorporate the most significant outcomes into one, easy-to-use clinical determinant.
Collapse
Affiliation(s)
- Maxim Van Herreweghe
- Natural Products and Food Research and Analysis-Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Tess De Bruyne
- Natural Products and Food Research and Analysis-Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Nina Hermans
- Natural Products and Food Research and Analysis-Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Ralph Huits
- Department of Infectious Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Negrar, Italy
| |
Collapse
|
14
|
Vianna CM, Noronha TG, Camacho LAB, Andrade RC, de Souza Brum RC, Dos Santos EM, Aguiar DF, Dos Santos MLB, de Souza Cruz RL, de Lima SMB, de Souza Azevedo Soares A, Schwarcz WD, da Matta de Castro T, Xavier JR, da Conceição DA, Homma A, de Lourdes de Sousa Maia M. Duration of post-vaccination immunity to yellow fever in volunteers ten years after a dose-response study - A complementary study. Vaccine 2024; 42:126083. [PMID: 38926068 PMCID: PMC11413730 DOI: 10.1016/j.vaccine.2024.06.050] [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: 04/15/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
A single dose of standard yellow fever (YF) vaccine is considered to provide life-long protection. In this study, we evaluate the seropositivity conferred by lower doses 10 years post-vaccination. In 2009, Bio-Manguinhos/Fiocruz performed a dose-response study with the 17DD yellow fever vaccine, administering the vaccine in the usual mean dose of 27.476 IU and in decreasing doses (10.447 IU, 3.013 IU, 587 IU, 158 IU and 31 IU), with the usual volume and route (0,5 ml subcutaneous). The decreasing doses were obtained by dilution in the laboratory of the manufacturer and the lots in test had standard quality control and were produced by good manufacturing practices (GMP). Around 30 days after the vaccination, doses down to 587 IU had similar immunogenicity and the 158 IU and 31 IU were inferior to the full dose. The seropositivity was maintained for 10 months, except on the 31 IU group. Eight years after, 85 % of 318 participants evaluated in a follow-up, maintained seropositivity that was similar across groups. Consistently, antibody titers in the reduced-dose groups were also comparable to those of the full-dose group. The current study, 10 years later, showed similarity between the vaccine groups (six arms who received the YF vaccine in decreasing doses: 27.476 IU, 10.447 IU, 3.013 IU, 587 IU, 158 IU, 31 IU) both in relation of seropositivity and in the evaluation of the geometric mean titers. The seropositivity rates across subgroups were 83,1%, 90 %, 87 %, 93 %, 83,8% and 85 %, correspondingly. These findings provides further support to the long-term immunogenicity of lower doses. Clinical trial registry: NCT04416477.
Collapse
Affiliation(s)
- Clarice Monteiro Vianna
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil.
| | | | | | - Raissa Coelho Andrade
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Eliane Matos Dos Santos
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | | | | | | | | | - Waleska Dias Schwarcz
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Janaína Reis Xavier
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Akira Homma
- Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
15
|
Guo W, Jiang T, Rao J, Zhang Z, Zhang X, Su J, Yin C, Lu M, Hu X, Shan C. A safer cell-based yellow fever live attenuated vaccine protects mice against YFV infection. iScience 2024; 27:110972. [PMID: 39398246 PMCID: PMC11470684 DOI: 10.1016/j.isci.2024.110972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/07/2024] [Accepted: 09/13/2024] [Indexed: 10/15/2024] Open
Abstract
The live attenuated yellow fever vaccine (YF17D) has caused controversial safety issues in history with low-yield problems, which has led to a large population being unable to be vaccinated and vaccine shortage in facing recent outbreaks. Here, we report a safer live attenuated vaccine candidate, YF17D-Δ77, which contains 77 nucleotides deletion in the 3' untranslated region (3' UTR) of the YF17D genome. YF17D-Δ77 exhibited no neurotropism and decreased viscerotropism and caused significantly lower lethality in mice compared to YF17D. Mechanistically, the deletion enhanced the sensitivity of the virus to type I and type II interferon responses, which hindered viral replication. Encouragingly, YF17D-Δ77 provided comparable immune protection in mice as did YF17D. Even 10 PFU of YF17D-Δ77 completely protected mice against YFV-Asibi challenge. In addition, the Δ77 mutation showed excellent stability after successive passages in Vero cells. Collectively, the data suggest that further development of YF17D-Δ77 as vaccine candidate is warranted.
Collapse
Affiliation(s)
- Weiwei Guo
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Tingting Jiang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Juhong Rao
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Zihan Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xuekai Zhang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Jiaoling Su
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Chunhong Yin
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Mingqing Lu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
| | - Xue Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
| | - Chao Shan
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, China
- University of the Chinese Academy of Sciences, Beijing 100039, China
- Hubei Jiangxia Laboratory, Wuhan 430200, China
| |
Collapse
|
16
|
Shanshin DV, Borisevich SS, Shaprova ON, Nesmeyanova VS, Bondar AA, Porozov YB, Khamitov EM, Kolosova EA, Shelemba AA, Ushkalenko ND, Protopopova EV, Sergeev AA, Loktev VB, Shcherbakov DN. Phage Display Revealed the Complex Structure of the Epitope of the Monoclonal Antibody 10H10. Int J Mol Sci 2024; 25:10311. [PMID: 39408641 PMCID: PMC11476565 DOI: 10.3390/ijms251910311] [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: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 10/20/2024] Open
Abstract
The annual number of reported human cases of flavivirus infections continues to increase. Measures taken by local healthcare systems and international organizations are not fully successful. In this regard, new approaches to treatment and prevention of flavivirus infections are relevant. One promising approach is to use monoclonal antibody preparations. The mouse mAb 10H10 is capable of interacting with viruses belonging to the genus Orthoflavivirus which are pathogenic to humans. ELISA and molecular modeling data can indicate that mAb 10H10 recognizes the fusion loop region of E protein. The KD of interaction between the mAb 10H10 and recombinant analogs of the E protein of the tick-borne encephalitis (TBEV), Zika (ZIKV) and dengue (DENV) viruses range from 1.5 to 4 nM. The aim of this study was to map the epitope of this antibody using phage display technology. After three rounds of biopanning, 60 individual phage clones were chosen. The amino acid sequences of the selected peptides were conveniently divided into five groups. Based on the selected peptides, bacteriophages were obtained carrying peptides on the surfaces of the pIII and pVIII proteins, which were tested for binding to the antibody in ELISA. Thus, the epitope of the mAb 10H10 is the highly conserved region 98-DRGWGNXXGLFGK-110 of the flavivirus E protein. The structures of the complexes of the identified peptides with the antibody paratope are proposed using the molecular docking and dynamics methods.
Collapse
Affiliation(s)
- Daniil V. Shanshin
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Sophia S. Borisevich
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
- Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, Koltsovo 630559, Russia
| | - Olga N. Shaprova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Valentina S. Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Alexander A. Bondar
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia;
| | - Yuri B. Porozov
- Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia;
- Advitam Laboratory, 11108 Belgrade, Serbia
| | - Edward M. Khamitov
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
| | - Evgeniia A. Kolosova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Arseniya A. Shelemba
- Federal State Budgetary Scientific Institution “Federal Research Center for Fundamental and Translational Medicine”, Novosibirsk 630117, Russia;
| | - Nikita D. Ushkalenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Elena V. Protopopova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Artemiy A. Sergeev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Valery B. Loktev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Dmitriy N. Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| |
Collapse
|
17
|
Muccilli SG, Schwarz B, Jessop F, Shannon JG, Bohrnsen E, Shue B, Hong SH, Hsu T, Ashbrook AW, Guarnieri JW, Lack J, Wallace DC, Bosio CM, MacDonald MR, Rice CM, Yewdell JW, Best SM. Mitochondrial Hyperactivity and Reactive Oxygen Species Drive Innate Immunity to the Yellow Fever Virus-17D Live-Attenuated Vaccine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611167. [PMID: 39282299 PMCID: PMC11398391 DOI: 10.1101/2024.09.04.611167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The yellow fever virus 17D (YFV-17D) live attenuated vaccine is considered one of the successful vaccines ever generated associated with high antiviral immunity, yet the signaling mechanisms that drive the response in infected cells are not understood. Here, we provide a molecular understanding of how metabolic stress and innate immune responses are linked to drive type I IFN expression in response to YFV-17D infection. Comparison of YFV-17D replication with its parental virus, YFV-Asibi, and a related dengue virus revealed that IFN expression requires RIG-I-like Receptor signaling through MAVS, as expected. However, YFV-17D uniquely induces mitochondrial respiration and major metabolic perturbations, including hyperactivation of electron transport to fuel ATP synthase. Mitochondrial hyperactivity generates reactive oxygen species (mROS) and peroxynitrite, blocking of which abrogated IFN expression in non-immune cells without reducing YFV-17D replication. Scavenging ROS in YFV-17D-infected human dendritic cells increased cell viability yet globally prevented expression of IFN signaling pathways. Thus, adaptation of YFV-17D for high growth uniquely imparts mitochondrial hyperactivity generating mROS and peroxynitrite as the critical messengers that convert a blunted IFN response into maximal activation of innate immunity essential for vaccine effectiveness.
Collapse
Affiliation(s)
- Samantha G. Muccilli
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD
| | | | - Forrest Jessop
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, MT
| | - Jeffrey G. Shannon
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT
| | - Eric Bohrnsen
- Research Technologies Branch, NIAID, NIH, Hamilton, MT
| | - Byron Shue
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT
| | - Seon-Hui Hong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Thomas Hsu
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT
| | - Alison W. Ashbrook
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, NIAID, NIH
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, MT
| | - Margaret R. MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Jonathan W. Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD
| | - Sonja M. Best
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, MT
| |
Collapse
|
18
|
Giannakopoulou E, Akrani I, Mpekoulis G, Frakolaki E, Dimitriou M, Myrianthopoulos V, Vassilaki N, Zoidis G. Novel Pyrazino[1,2- a]indole-1,3(2 H,4 H)-dione Derivatives Targeting the Replication of Flaviviridae Viruses: Structural and Mechanistic Insights. Viruses 2024; 16:1238. [PMID: 39205212 PMCID: PMC11360281 DOI: 10.3390/v16081238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Infections with Flaviviridae viruses, such as hepatitis C (HCV), dengue (DENV), and yellow fever (YFV) viruses, are major public health problems worldwide. In the case of HCV, treatment is associated with drug resistance and high costs, while there is no clinically approved therapy for DENV and YFV. Consequently, there is still a need for new chemotherapies with alternative modes of action. We have previously identified novel 2-hydroxypyrazino[1,2-a]indole-1,3(2H,4H)-diones as metal-chelating inhibitors targeting HCV RNA replication. Here, by utilizing a structure-based approach, we rationally designed a second series of compounds by introducing various substituents at the indole core structure and at the imidic nitrogen, to improve specificity against the RNA-dependent RNA polymerase (RdRp). The resulting derivatives were evaluated for their potency against HCV genotype 1b, DENV2, and YFV-17D using stable replicon cell lines. The most favorable substitution was nitro at position 6 of the indole ring (compound 36), conferring EC50 1.6 μM against HCV 1b and 2.57 μΜ against HCV 1a, with a high selectivity index. Compound 52, carrying the acetohydroxamic acid functionality (-CH2CONHOH) on the imidic nitrogen, and compound 78, the methyl-substituted molecule at the position 4 indolediketopiperazine counterpart, were the most effective against DENV and YFV, respectively. Interestingly, compound 36 had a high genetic barrier to resistance and only one resistance mutation was detected, T181I in NS5B, suggesting that the compound target HCV RdRp is in accordance with our predicted model.
Collapse
Affiliation(s)
- Erofili Giannakopoulou
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece; (E.G.); (I.A.); (V.M.)
| | - Ifigeneia Akrani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece; (E.G.); (I.A.); (V.M.)
| | - George Mpekoulis
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, GR-11521 Athens, Greece; (G.M.); (M.D.)
| | - Efseveia Frakolaki
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, GR-11521 Athens, Greece; (G.M.); (M.D.)
| | - Marios Dimitriou
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, GR-11521 Athens, Greece; (G.M.); (M.D.)
| | - Vassilios Myrianthopoulos
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece; (E.G.); (I.A.); (V.M.)
| | - Niki Vassilaki
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, GR-11521 Athens, Greece; (G.M.); (M.D.)
| | - Grigoris Zoidis
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimiopolis-Zografou, GR-15771 Athens, Greece; (E.G.); (I.A.); (V.M.)
| |
Collapse
|
19
|
Pan Q, Chen Q, Zhang W, Jiao H, Yu L, Hu H. Structural Insights into the Dynamic Assembly of a YFV sNS1 Tetramer. Viruses 2024; 16:1212. [PMID: 39205186 PMCID: PMC11359903 DOI: 10.3390/v16081212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 09/04/2024] Open
Abstract
Yellow fever virus (YFV) infections can cause severe diseases in humans, resulting in mass casualties in Africa and the Americas each year. Secretory NS1 (sNS1) is thought to be used as a diagnostic marker of flavivirus infections, playing an essential role in the flavivirus life cycle, but little is known about the composition and structure of YFV sNS1. Here, we present that the recombinant YFV sNS1 exists in a heterogeneous mixture of oligomerizations, predominantly in the tetrameric form. The cryoEM structures show that the YFV tetramer of sNS1 is stacked by the hydrophobic interaction between β-roll domains and greasy fingers. According to the 3D variability analysis, the tetramer is in a semi-stable state that may contain multiple conformations with dynamic changes. We believe that our study provides critical insights into the oligomerization of NS1 and will aid the development of NS1-based diagnoses and therapies.
Collapse
Affiliation(s)
- Qi Pan
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Qiang Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Wanqin Zhang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Haizhan Jiao
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| | - Lei Yu
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510060, China
| | - Hongli Hu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China; (Q.P.); (Q.C.); (W.Z.); (H.J.)
| |
Collapse
|
20
|
Chen Y, Li Y, Lu L, Zou P. Zafirlukast, as a viral inactivator, potently inhibits infection of several flaviviruses, including Zika virus, dengue virus, and yellow fever virus. Antimicrob Agents Chemother 2024; 68:e0016824. [PMID: 38809067 PMCID: PMC11232407 DOI: 10.1128/aac.00168-24] [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: 01/31/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
Zika virus (ZIKV) is one of the mosquito-borne flaviviruses that exhibits a unique tropism to nervous systems and is associated with Guillain-Barre syndrome and congenital Zika syndrome (CZS). Dengue virus (DENV) and yellow fever virus (YFV), the other two mosquito-borne flaviviruses, have also been circulating for a long time and cause severe diseases, such as dengue hemorrhagic fever and yellow fever, respectively. However, there are no safe and effective antiviral drugs approved for the treatment of infections or coinfections of these flaviviruses. Here, we found that zafirlukast, a pregnancy-safe leukotriene receptor antagonist, exhibited potent antiviral activity against infections of ZIKV strains from different lineages in different cell lines, as well as against infections of DENV-2 and YFV 17D. Mechanistic studies demonstrated that zafirlukast directly and irreversibly inactivated these flaviviruses by disrupting the integrity of the virions, leading to the loss of viral infectivity, hence inhibiting the entry step of virus infection. Considering its efficacy against flaviviruses, its safety for pregnant women, and its neuroprotective effect, zafirlukast is a promising candidate for prophylaxis and treatment of infections or coinfections of ZIKV, DENV, and YFV, even in pregnant women.
Collapse
Affiliation(s)
- Yongkang Chen
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuan Li
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lu Lu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Mensah EA, Gyasi SO, Nsubuga F, Alali WQ. A proposed One Health approach to control yellow fever outbreaks in Uganda. ONE HEALTH OUTLOOK 2024; 6:9. [PMID: 38783349 PMCID: PMC11119388 DOI: 10.1186/s42522-024-00103-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Yellow Fever (YF) is an acute viral hemorrhagic disease. Uganda is located within the Africa YF belt. Between 2019 and 2022, the Ugandan Health Authorities reported at least one outbreak of YF annually with an estimated 892 suspected cases, on average per year. The persistent recurrence of this disease raises significant concerns about the efficacy of current response strategies and prevention approaches. YF has been recognized as a One Health issue due to its interrelatedness with the animal and environmental domains. Monkeys have been recognized as the virus primary reservoir. The YF virus is transmitted through bites of infected Aedes or Haemagogus species mosquitoes between monkeys and humans. Human activities, monkey health, and environmental health issues (e.g., climate change and land use) impact YF incidence in Uganda. Additionally, disease control programs for other tropical diseases, such as mosquitoes control programs for malaria, impact YF incidence.This review adopts the One Health approach to highlight the limitations in the existing segmented YF control and prevention strategies in Uganda, including the limited health sector surveillance, the geographically localized outbreak response efforts, the lack of a comprehensive vaccination program, the limited collaboration and communication among relevant national and international agencies, and the inadequate vector control practices. Through a One Health approach, we propose establishing a YF elimination taskforce. This taskforce would oversee coordination of YF elimination initiatives, including implementing a comprehensive surveillance system, conducting mass YF vaccination campaigns, integrating mosquito management strategies, and enhancing risk communication. It is anticipated that adopting the One Health approach will reduce the risk of YF incidence and outbreaks.
Collapse
Affiliation(s)
- Emmanuel Angmorteh Mensah
- Department of Biostatistics & Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA
| | - Samuel Ofori Gyasi
- Department of Immunization, Vaccines and Biologicals, World Health Organization Country Office, Kampala, Uganda
| | - Fred Nsubuga
- Division of Immunization and Vaccines, Ministry of Health, Kampala, Uganda
| | - Walid Q Alali
- Department of Biostatistics & Epidemiology, College of Public Health, East Tennessee State University, Johnson City, TN, USA.
| |
Collapse
|
22
|
Gonçalves AP, Almeida LT, de Rezende IM, Fradico JRB, Pereira LS, Ramalho DB, Pascoal Xavier MA, Calzavara Silva CE, Monath TP, LaBeaud AD, Drumond BP, Campi-Azevedo AC, Martins-Filho OA, Teixeira-Carvalho A, Alves PA, Grupo de Estudos de Pesquisa e Resposta em Febre Amarela do Estado de Minas Gerais. Evaluation of humoral immune response after yellow fever infection: an observational study on patients from the 2017-2018 sylvatic outbreak in Brazil. Microbiol Spectr 2024; 12:e0370323. [PMID: 38511952 PMCID: PMC11064539 DOI: 10.1128/spectrum.03703-23] [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: 10/17/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Between 2016 and 2018, Brazil experienced major sylvatic yellow fever (YF) outbreaks that caused hundreds of casualties, with Minas Gerais (MG) being the most affected state. These outbreaks provided a unique opportunity to assess the immune response triggered by the wild-type (WT) yellow fever virus (YFV) in humans. The plaque reduction neutralization test (PRNT) is currently the standard method to assess the humoral immune response to YFV by measuring neutralizing antibodies (nAbs). The present study aimed to evaluate the humoral immune response of patients from the 2017-2018 sylvatic YF outbreak in MG with different disease outcomes by using PRNTs with a WT YFV strain, isolated from the 2017-2018 outbreak, and a vaccine YFV strain. Samples from naturally infected YF patients were tested, in comparison with healthy vaccinees. Results showed that both groups presented different levels of nAb against the WT and vaccine strains, and the levels of neutralization against the strains varied homotypically and heterotypically. Results based on the geometric mean titers (GMTs) suggest that the humoral immune response after a natural infection of YFV can reach higher levels than that induced by vaccination (GMT of patients against WT YFV compared to GMT of vaccinees, P < 0.0001). These findings suggest that the humoral immune responses triggered by the vaccine and WT strains of YFV are different, possibly due to genetic and antigenic differences between these viruses. Therefore, current means of assessing the immune response in naturally infected YF individuals and immunological surveillance methods in areas with intense viral circulation may need to be updated.IMPORTANCEYellow fever is a deadly febrile disease caused by the YFV. Despite the existence of effective vaccines, this disease still represents a public health concern worldwide. Much is known about the immune response against the vaccine strains of the YFV, but recent studies have shown that it differs from that induced by WT strains. The extent of this difference and the mechanisms behind it are still unclear. Thus, studies aimed to better understand the immune response against this virus are relevant and necessary. The present study evaluated levels of neutralizing antibodies of yellow fever patients from recent outbreaks in Brazil, in comparison with healthy vaccinees, using plaque reduction neutralization tests with WT and vaccine YFV strains. Results showed that the humoral immune response in naturally infected patients was higher than that induced by vaccination, thus providing new insights into the immune response triggered against these viruses.
Collapse
Affiliation(s)
| | - Letícia Trindade Almeida
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Izabela Maurício de Rezende
- Department of Pediatrics, Infectious Disease Division, Stanford University School of Medicine, Stanford, California, USA
| | | | - Leonardo Soares Pereira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
| | - Dario Brock Ramalho
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Antônio Pascoal Xavier
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
- Departamento de Anatomia Patológica e Medicina Legal, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Angelle Desiree LaBeaud
- Department of Pediatrics, Infectious Disease Division, Stanford University School of Medicine, Stanford, California, USA
| | - Betania Paiva Drumond
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Andréa Teixeira-Carvalho
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Pedro Augusto Alves
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
| | - Grupo de Estudos de Pesquisa e Resposta em Febre Amarela do Estado de Minas Gerais
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, Minas Gerais, Brazil
- Department of Pediatrics, Infectious Disease Division, Stanford University School of Medicine, Stanford, California, USA
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
- Departamento de Anatomia Patológica e Medicina Legal, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Crozet BioPharma LLC, Lexington, Massachusetts, USA
| |
Collapse
|
23
|
Saidu JZ, Eghafona NO. Circulation of yellow fever virus in Benin city, Edo state Nigeria. Biologicals 2024; 86:101765. [PMID: 38593685 DOI: 10.1016/j.biologicals.2024.101765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 12/07/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Yellow fever (YF) is one of the most acute viral hemorrhagic diseases of the 18th and 19th centuries, which continues to cause severe morbidity and mortality in Africa. After 21 years of no reported cases of yellow fever in Nigeria, till 2017 where a case was confirmed in Kwara State, also in November 2018,WHO was informed of a cluster of suspected yellow fever cases and deaths in Edo state, Nigeria. The study was among all age group attending health centres in Benin City, Edo state. A total of 280 blood samples were collected from consented febrile patients and were screened for antibodies to Zika virus using rapid diagnostic test (RDT) kits. Blood samples positive to Zika virus (IgM/IgG RDT), were subjected to molecular characterization. Using the flavividae family primers, six (6) samples where confirmed positive by Hemi-nested reverse transcription PCR (hnRT-PCR) sequencing. Nucleotide sequence blast revealed the sequenceswere similar to Yellow fever virus strains. Phylogenetic analysis revealed that the yellow fever virus sequences are closely related to the African strains. Despite the safe and effective yellow fever vaccine, yellow fever virus is seen to be in circulation, hence the need for continues mass vaccination.
Collapse
Affiliation(s)
- Joy Zitgwai Saidu
- Department of Microbiology, Faculty of Life Sciences, University of Benin, PMB 1154, Benin City, Edo State, Nigeria.
| | - Nosakhare Odeh Eghafona
- Department of Microbiology, Faculty of Life Sciences, University of Benin, PMB 1154, Benin City, Edo State, Nigeria; Department of Microbiology, Faculty of Sciences, Benson, Idahosa University, Benin City, Nigeria
| |
Collapse
|
24
|
Huebl L, Nnyombi A, Kihumuro A, Lukwago D, Walakira E, Kutalek R. Perceptions of yellow fever emergency mass vaccinations among vulnerable groups in Uganda: A qualitative study. PLoS Negl Trop Dis 2024; 18:e0012173. [PMID: 38739650 PMCID: PMC11115279 DOI: 10.1371/journal.pntd.0012173] [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: 02/10/2023] [Revised: 05/23/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Yellow fever (YF), a mosquito-borne viral hemorrhagic fever, is endemic in Uganda and causes frequent outbreaks. A total of 1.6 million people were vaccinated during emergency mass immunization campaigns in 2011 and 2016. This study explored local perceptions of YF emergency mass immunization among vulnerable groups to inform future vaccination campaigns. METHODOLOGY In this qualitative study, we conducted 43 semi-structured interviews, 4 focus group discussions, and 10 expert interviews with 76 participants. Data were collected in six affected districts with emergency mass vaccination. We included vulnerable groups (people ≥ 65 years and pregnant women) who are typically excluded from YF vaccination except during mass immunization. Data analysis was conducted using grounded theory. Inductive coding was utilized, progressing through open, axial, and selective coding. PRINCIPAL FINDINGS Participants relied on community sources for information about the YF mass vaccination. Information was disseminated door-to-door, in community spaces, during religious gatherings, and on the radio. However, most respondents had no knowledge of the vaccine, and it was unclear to them whether a booster dose was required. In addition, the simultaneous presidential election during the mass vaccination campaign led to suspicion and resistance to vaccination. The lack of reliable and trustworthy information and the politicization of vaccination campaigns reinforced mistrust of YF vaccines. CONCLUSIONS/SIGNIFICANCE People in remote areas affected by YF outbreaks rely on community sources of information. We therefore recommend improving health education, communication, and engagement through respected and trusted community members. Vaccination campaigns can never be seen as detached from political systems and power relations.
Collapse
Affiliation(s)
- Lena Huebl
- Unit Medical Anthropology and Global Health, Department of Social and Preventive Medicine, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine & I Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Aloysious Nnyombi
- Department of Social Work and Social Administration, Makerere University, Kampala, Uganda
| | - Aban Kihumuro
- Department of Nursing and Health Sciences, Bishop Stuart University, Mbarara, Uganda
| | - Denis Lukwago
- Cluster Monitoring and Evaluation Lead, Rakai Health Sciences Program, Masaka, Uganda
| | - Eddy Walakira
- Department of Social Work and Social Administration, Makerere University, Kampala, Uganda
| | - Ruth Kutalek
- Unit Medical Anthropology and Global Health, Department of Social and Preventive Medicine, Center for Public Health, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Reis LR, Costa-Rocha IA, Abdala-Torres T, Campi-Azevedo AC, Peruhype-Magalhães V, Araújo MSS, Spezialli E, do Valle Antonelli LR, da Silva-Pereira RA, Almeida GG, Fernandes EG, Fantinato FFST, Domingues CMAS, Lemos MCF, Chieppe A, Lemos JAC, Coelho-Dos-Reis JG, de Lima SMB, de Souza Azevedo A, Schwarcz WD, Camacho LAB, de Lourdes de Sousa Maia M, de Noronha TG, Duault C, Rosenberg-Hasson Y, Teixeira-Carvalho A, Maecker HT, Martins-Filho OA. Comprehensive landscape of neutralizing antibody and cell-mediated response elicited by the 1/5 fractional dose of 17DD-YF primary vaccination in adults. Sci Rep 2024; 14:7709. [PMID: 38565882 PMCID: PMC10987530 DOI: 10.1038/s41598-024-57645-3] [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: 10/11/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
The present study aimed at evaluating the YF-specific neutralizing antibody profile besides a multiparametric analysis of phenotypic/functional features of cell-mediated response elicited by the 1/5 fractional dose of 17DD-YF vaccine, administered as a single subcutaneous injection. The immunological parameters of each volunteer was monitored at two time points, referred as: before (Day 0) [Non-Vaccinated, NV(D0)] and after vaccination (Day 30-45) [Primary Vaccinees, PV(D30-45)]. Data demonstrated high levels of neutralizing antibodies for PV(D30-45) leading to a seropositivity rate of 93%. A broad increase of systemic soluble mediators with a mixed profile was also observed for PV(D30-45), with IFN-γ and TNF-α presenting the highest baseline fold changes. Integrative network mapping of soluble mediators showed increased correlation numbers in PV(D30-45) as compared to NV(D0) (532vs398). Moreover, PV(D30-45) exhibited increased levels of Terminal Effector (CD45RA+CCR7-) CD4+ and CD8+ T-cells and Non-Classical memory B-cells (IgD+CD27+). Dimensionality reduction of Mass Cytometry data further support these findings. A polyfunctional cytokine profile (TNF-α/IFN-γ/IL-10/IL-17/IL-2) of T and B-cells was observed upon in vitro antigen recall. Mapping and kinetics timeline of soluble mediator signatures for PV(D30-45) further confirmed the polyfunctional profile upon long-term in vitro culture, mediated by increased levels of IFN-γ and TNF-α along with decreased production of IL-10. These findings suggest novel insights of correlates of protection elicited by the 1/5 fractional dose of 17DD-YF vaccine.
Collapse
Affiliation(s)
- Laise Rodrigues Reis
- Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | | | - Thais Abdala-Torres
- Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | - Elaine Spezialli
- Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | | | | | - Alexandre Chieppe
- Superintendência de Vigilância em Saúde, Secretaria Municipal de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Jordana Grazziela Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais - UFMG, Belo Horizonte, MG, Brazil
| | - Sheila Maria Barbosa de Lima
- Departamento de Desenvolvimento Experimental e Pré-Clínico, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Adriana de Souza Azevedo
- Laboratório de Análise Imunomolecular, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Waleska Dias Schwarcz
- Laboratório de Análise Imunomolecular, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | | | - Tatiana Guimarães de Noronha
- Assessoria Clínica, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos - FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Caroline Duault
- Human Immune Monitoring Center, Stanford University, Stanford, CA, USA
| | | | | | - Holden Terry Maecker
- Human Immune Monitoring Center, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, USA.
| | - Olindo Assis Martins-Filho
- Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, MG, Brazil.
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, MG, Brazil.
| |
Collapse
|
26
|
Abdala-Torres T, Campi-Azevedo AC, da Silva-Pereira RA, Dos Santos LI, Henriques PM, Costa-Rocha IA, Otta DA, Peruhype-Magalhães V, Teixeira-Carvalho A, Araújo MSS, Fernandes EG, Sato HK, Fantinato FFST, Domingues CMAS, Kallás EG, Tomiyama HTI, Lemos JAC, Coelho-Dos-Reis JG, de Lima SMB, Schwarcz WD, de Souza Azevedo A, Trindade GF, Ano Bom APD, da Silva AMV, Fernandes CB, Camacho LAB, de Sousa Maia MDL, Martins-Filho OA, do Antonelli LRDV. Immune response induced by standard and fractional doses of 17DD yellow fever vaccine. NPJ Vaccines 2024; 9:54. [PMID: 38459059 PMCID: PMC10923915 DOI: 10.1038/s41541-024-00836-w] [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: 10/17/2023] [Accepted: 02/05/2024] [Indexed: 03/10/2024] Open
Abstract
The re-emergence of yellow fever (YF) urged new mass vaccination campaigns and, in 2017, the World Health Organization approved the use of the fractional dose (FD) of the YF vaccine due to stock shortage. In an observational cross-sectional investigation, we have assessed viremia, antibodies, soluble mediators and effector and memory T and B-cells induced by primary vaccination of volunteers with FD and standard dose (SD). Similar viremia and levels of antibodies and soluble markers were induced early after immunization. However, a faster decrease in the latter was observed after SD. The FD led to a sustained expansion of helper T-cells and an increased expression of activation markers on T-cells early after vaccination. Although with different kinetics, expansion of plasma cells was induced upon SD and FD immunization. Integrative analysis reveals that FD induces a more complex network involving follicular helper T cells and B-cells than SD. Our findings substantiate that FD can replace SD inducing robust correlates of protective immune response against YF.
Collapse
Affiliation(s)
- Thais Abdala-Torres
- Laboratório de Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Carolina Campi-Azevedo
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | - Rosiane Aparecida da Silva-Pereira
- Laboratório de Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | | | - Priscilla Miranda Henriques
- Laboratório de Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | - Ismael Artur Costa-Rocha
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | - Dayane Andriotti Otta
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | - Vanessa Peruhype-Magalhães
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil
| | | | - Eder Gatti Fernandes
- Divisão de Imunização, Secretaria de Estado de Saúde de São Paulo, São Paulo, SP, Brazil
- Departamento de Vigilância das Doenças Transmissíveis, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, DF, Brazil
| | - Helena Keico Sato
- Divisão de Imunização, Secretaria de Estado de Saúde de São Paulo, São Paulo, SP, Brazil
| | | | | | - Esper Georges Kallás
- Departamento de Doenças Infecciosas e Parasitárias, Escola de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil
| | | | | | - Jordana Grazziela Coelho-Dos-Reis
- Laboratório de Virologia Básica e Aplicada, Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Sheila Maria Barbosa de Lima
- Departamento de Desenvolvimento Experimental e Pré-clínico, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Waleska Dias Schwarcz
- Laboratório de Análise Imunomecular, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Adriana de Souza Azevedo
- Laboratório de Análise Imunomecular, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Gisela Freitas Trindade
- Laboratório de Tecnologia Virológica, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Ana Paula Dinis Ano Bom
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Andrea Marques Vieira da Silva
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Camilla Bayma Fernandes
- Laboratório de Tecnologia Imunológica, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | | | - Maria de Lourdes de Sousa Maia
- Departamento de Assuntos Médicos, Estudos Clínicos e Vigilância Pós-Registro, Instituto de Tecnologia em Imunobiológicos Bio-Manguinhos, FIOCRUZ, Rio de Janeiro, RJ, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil.
| | - Lis Ribeiro do Valle do Antonelli
- Laboratório de Biologia e Imunologia de Doenças Infecciosas e Parasitárias, Instituto René Rachou, FIOCRUZ-Minas, Belo Horizonte, MG, Brazil.
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
27
|
Coelho Ferraz A, Bueno da Silva Menegatto M, Lameira Souza Lima R, Samuel Ola-Olub O, Caldeira Costa D, Carlos de Magalhães J, Maurício Rezende I, Desiree LaBeaud A, P Monath T, Augusto Alves P, Teixeira de Carvalho A, Assis Martins-Filho O, P Drumond B, Magalhães CLDB. Yellow fever virus infection in human hepatocyte cells triggers an imbalance in redox homeostasis with increased reactive oxygen species production, oxidative stress, and decreased antioxidant enzymes. Free Radic Biol Med 2024; 213:266-273. [PMID: 38278309 PMCID: PMC10911966 DOI: 10.1016/j.freeradbiomed.2024.01.042] [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] [Received: 11/19/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 01/28/2024]
Abstract
Yellow fever (YF) presents a wide spectrum of severity, with clinical manifestations in humans ranging from febrile and self-limited to fatal cases. Although YF is an old disease for which an effective and safe vaccine exists, little is known about the viral- and host-specific mechanisms that contribute to liver pathology. Several studies have demonstrated that oxidative stress triggered by viral infections contributes to pathogenesis. We evaluated whether yellow fever virus (YFV), when infecting human hepatocytes cells, could trigger an imbalance in redox homeostasis, culminating in oxidative stress. YFV infection resulted in a significant increase in reactive oxygen species (ROS) levels from 2 to 4 days post infection (dpi). When measuring oxidative parameters at 4 dpi, YFV infection caused oxidative damage to lipids, proteins, and DNA, evidenced by an increase in lipid peroxidation/8-isoprostane, carbonyl protein, and 8-hydroxy-2'-deoxyguanosine, respectively. Furthermore, there was a significant reduction in the activity of the antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx), in addition to a reduction in the ratio of reduced to oxidized glutathione (GSH/GSSG), indicating a pro-oxidant environment. However, no changes were observed in the enzymatic activity of the enzyme catalase (CAT) or in the gene expression of SOD isoforms (1/2/3), CAT, or GPx. Therefore, our results show that YFV infection generates an imbalance in redox homeostasis, with the overproduction of ROS and depletion of antioxidant enzymes, which induces oxidative damage to cellular constituents. Moreover, as it has been demonstrated that oxidative stress is a conspicuous event in YFV infection, therapeutic strategies based on antioxidant biopharmaceuticals may be new targets for the treatment of YF.
Collapse
Affiliation(s)
- Ariane Coelho Ferraz
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Marília Bueno da Silva Menegatto
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Rafaela Lameira Souza Lima
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Oluwashola Samuel Ola-Olub
- Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Daniela Caldeira Costa
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - José Carlos de Magalhães
- Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Departamento de Química, Biotecnologia e Engenharia de Bioprocessos, Universidade Federal de São João del-Rei, Ouro Branco, Minas Gerais, Brazil
| | - Izabela Maurício Rezende
- Pandemic Preparedenss Hub, Divison of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Angelle Desiree LaBeaud
- Division of Infectious Diseases, Department of Pediatrics, Stanford University School of Medicine, California, United States
| | | | - Pedro Augusto Alves
- Imunologia de Doenças Virais, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira de Carvalho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisa em Biomarcadores, Instituto René Rachou, Fundação Oswaldo Cruz - FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Betânia P Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Cintia Lopes de Brito Magalhães
- Programa de Pós-Graduação em Ciências Biológicas, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil; Programa de Pós-Graduação em Biotecnologia, Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, Ouro Preto, Minas Gerais, Brazil.
| |
Collapse
|
28
|
Garcia-Oliveira GF, Guimarães ACDS, Moreira GD, Costa TA, Arruda MS, de Mello ÉM, Silva MC, de Almeida MG, Hanley KA, Vasilakis N, Drumond BP. YELLOW ALERT: Persistent Yellow Fever Virus Circulation among Non-Human Primates in Urban Areas of Minas Gerais State, Brazil (2021-2023). Viruses 2023; 16:31. [PMID: 38257732 PMCID: PMC10818614 DOI: 10.3390/v16010031] [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: 11/30/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Yellow fever virus (YFV) is the agent of yellow fever (YF), which affects both humans and non-human primates (NHP). Neotropical NHP are highly susceptible to YFV and considered sentinels for YFV circulation. Brazil faced a significant YF outbreak in 2017-2018, with over 2000 human cases and 2000 epizootics cases, mainly in the State of Minas Gerais, Brazil. This study aimed to investigate whether YFV circulation persisted in NHP after the human outbreak had subsided. To this end, NHP carcass samples collected in Minas Gerais from 2021 to 2023 were screened for YFV. RNA was extracted from tissue fragments and used in RT-qPCR targeting the YFV 5'UTR. Liver and lung samples from 166 animals were tested, and the detection of the β-actin mRNA was used to ensure adequacy of RNA isolation. YFV RNA was detected in the liver of 18 NHP carcasses collected mainly from urban areas in 2021 and 2022. YFV positive NHP were mostly represented by Callithrix, from 5 out of the 12 grouped municipalities (mesoregions) in Minas Gerais state. These findings reveal the continued YFV circulation in NHP in urban areas of Minas Gerais during 2021 and 2022, with the attendant risk of re-establishing the urban YFV cycle.
Collapse
Affiliation(s)
- Gabriela F. Garcia-Oliveira
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| | - Anna Catarina Dias Soares Guimarães
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| | - Gabriel Dias Moreira
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| | - Thais Alkifeles Costa
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| | - Matheus Soares Arruda
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| | - Érica Munhoz de Mello
- Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte CEP 31270-705, Minas Gerais, Brazil
| | - Marlise Costa Silva
- Laboratório de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte CEP 31270-705, Minas Gerais, Brazil
| | | | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, NM 88003-8801, USA;
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
- Center for Vector-Borne and Zoonotic Diseases, The University of Texas Medical Branch, Galveston, TX 77555-0609, USA
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, TX 77555-0610, USA
| | - Betânia Paiva Drumond
- Laboratório de Vírus, Departament of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte CEP 31270-901, Brazil; (G.F.G.-O.); (A.C.D.S.G.); (G.D.M.); (T.A.C.); (M.S.A.)
| |
Collapse
|
29
|
Zina SM, Hoarau G, Labetoulle M, Khairallah M, Rousseau A. Ocular Manifestations of Flavivirus Infections. Pathogens 2023; 12:1457. [PMID: 38133340 PMCID: PMC10747099 DOI: 10.3390/pathogens12121457] [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: 10/25/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
Flaviviruses are a group of positive-sense, single-stranded RNA viruses predominantly transmitted by arthropods (mainly mosquitoes) that cause severe endemic infections and epidemics on a global scale. They represent a major cause of systemic morbidity and death and are expanding worldwide. Among this group, dengue fever, the West Nile virus, yellow fever, Japanese Encephalitis, and, recently, the Zika virus have been linked to a spectrum of ocular manifestations. These manifestations encompass subconjunctival hemorrhages and conjunctivitis, anterior and posterior uveitis (inclusive of vitritis, chorioretinitis, and retinal vasculitis), maculopathy, retinal hemorrhages, and optic neuritis. Clinical diagnosis of these infectious diseases is primarily based on epidemiological data, history, systemic symptoms and signs, and the pattern of ocular involvement. Diagnosis confirmation relies on laboratory testing, including RT-PCR and serological testing. Ocular involvement typically follows a self-limited course but can result in irreversible visual impairment. Effective treatments of flavivirus infections are currently unavailable. Prevention remains the mainstay for arthropod vector and zoonotic disease control. Effective vaccines are available only for the yellow fever virus, dengue virus, and Japanese Encephalitis virus. This review comprehensively summarizes the current knowledge regarding the ophthalmic manifestations of the foremost flavivirus-associated human diseases.
Collapse
Affiliation(s)
- Sourour Meziou Zina
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Department of Ophthalmology, Faculty of Medicine, University of Monastir, Monastir 5019, Tunisia;
| | - Gautier Hoarau
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
| | - Marc Labetoulle
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Infectious Diseases Models for Innovative Therapies (IDMIT), French Alternative Energies and Atomic Commission (CEA), 92260 Fontenay-aux-Roses, France
| | - Moncef Khairallah
- Department of Ophthalmology, Faculty of Medicine, University of Monastir, Monastir 5019, Tunisia;
| | - Antoine Rousseau
- Department of Ophthalmology, Bicêtre Hospital, Public Assistance, Hospitals of Paris, Reference Network for Rare Diseases in Ophthalmology (OPHTARA), 94270 Le Kremlin-Bicêtre, France; (S.M.Z.); (G.H.); (M.L.)
- Center for Immunology of Viral, Auto-Immune, Hematological and Bacterial Diseases (IMVA-HB), Infectious Diseases Models for Innovative Therapies (IDMIT), French Alternative Energies and Atomic Commission (CEA), 92260 Fontenay-aux-Roses, France
| |
Collapse
|
30
|
Sura K, Rohilla H, Kumar D, Jakhar R, Ahlawat V, Kaushik D, Dangi M, Chhillar AK. Exploring structural antigens of yellow fever virus to design multi-epitope subunit vaccine candidate by utilizing an immuno-informatics approach. J Genet Eng Biotechnol 2023; 21:161. [PMID: 38051433 DOI: 10.1186/s43141-023-00621-7] [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: 04/21/2023] [Accepted: 11/15/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND Yellow fever is a mosquito-borne viral hemorrhagic disease transmitted by several species of virus-infected mosquitoes endemic to tropical regions of Central and South America and Africa. Earlier in the twentieth century, mass vaccination integrated with mosquito control was implemented to eradicate the yellow fever virus. However, regular outbreaks occur in these regions which pose a threat to travelers and residents of Africa and South America. There is no specific antiviral therapy, but there can be an effective peptide-based vaccine candidate to combat infection caused by the virus. Therefore, the study aims to design a multi-epitope-based subunit vaccine (MESV) construct against the yellow fever virus to reduce the time and cost using reverse vaccinology (RV) approach. METHODS Yellow fever virus contains 10,233 nucleotides that encode for 10 proteins (C, prM, E, NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) including 3 structural and 7 non-structural proteins. Structural proteins-precursor membrane protein (prM) and envelope protein (E)-were taken as a target for B cell and T cell epitope screening. Further, various immunoinformatics approaches were employed to FASTA sequences of structural proteins to retrieve B cell and T cell epitopes. MESV was constructed from these epitopes based on allergenicity, antigenicity and immunogenicity, toxicity, conservancy, and population coverage followed by structure prediction. The efficacy of the MESV construct to bind with human TLR-3, TLR-4, and TLR-8 were evaluated using molecular docking and simulation studies. Finally, in-silico cloning of vaccine construct was performed withpBR322 Escherichia coli expression system using codon optimization. RESULTS Predicted epitopes evaluated and selected for MESV construction were found stable, non-allergenic, highly antigenic, and global population coverage of 68.03% according to in-silico analysis. However, this can be further tested in in-vitro and in-vivo investigations. Epitopes were sequentially merged to construct a MESV consisting of 393 amino acids using adjuvant and linkers. Molecular docking and simulation studies revealed stable and high-affinity interactions. Furthermore, in-silico immune response graphs showed effective immune response generation. Finally, higher CAI value ensured high gene expression of vaccine in the host cell. CONCLUSION The designed MESV construct in the present in-silico study can be effective in generating an immune response against the yellow fever virus. Therefore, to prevent yellow fever, it can be an effective vaccine candidate. However, further downstream, in-vitro study is required.
Collapse
Affiliation(s)
- Kiran Sura
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Himanshi Rohilla
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Dev Kumar
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Ritu Jakhar
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
| | - Vaishali Ahlawat
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India
- Centre for Biotechnology, M.D. University, Rohtak, Haryana, India
| | | | - Mehak Dangi
- Centre for Bioinformatics, M.D. University, Rohtak, Haryana, India.
| | | |
Collapse
|
31
|
Ren H, Wang J, Tang H, Qian X, Xia B, Luo Z, Xu Z, Qi Z, Zhao P. Tiratricol inhibits yellow fever virus replication through targeting viral RNA-dependent RNA polymerase of NS5. Antiviral Res 2023; 219:105737. [PMID: 37879570 DOI: 10.1016/j.antiviral.2023.105737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/06/2023] [Accepted: 10/18/2023] [Indexed: 10/27/2023]
Abstract
Yellow fever virus (YFV) infection is a major public concern that threatens a large population in South America and Africa. No specific antiviral drugs are available for treating yellow fever. Here, we report that tiratricol (triiodothyroacetic acid, TRIAC), a clinically approved drug used to treat thyroid hormone resistance syndrome (THRS), is a potent YFV inhibitor both in host cells and in animal models.An in vitro study demonstrates that TRIAC remarkably suppresses viral RNA synthesis and protein expression in a dose-dependent manner in human hepatoma cell lines (Huh-7) with an EC50 value of 2.07 μM and a CC50 value of 385.77 μM respectively. The surface plasmon resonance assay and molecular docking analysis indicate that TRIAC hinders viral replication by binding to the RNA-dependent RNA polymerase (RdRp) domain of viral nonstructural protein NS5, probably through interacting with the active sites of RdRp.The inhibitory effect of TRIAC in vivo is also confirmed in 3-week old C57BL/6 mice challenged with YFV infection, from which the survival of the mice as well as lesions and infection in their tissues and serum issignificantly promoted following oral administration of TRIAC (0.2 mg/kg/day). Additionally, TRIAC shows a broad-spectrum antiviral activity against multiple flaviviruses such as TBEV, WNV,ZIKV, andJEV in vitro. Our data demonstrate that the TH analogue TRIAC is an effective anti-YFV compound and may act as a potential therapeutic candidate for the treatment of YFV infection if its clinical importance is determined in patients in future.
Collapse
Affiliation(s)
- Hao Ren
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Jiaqi Wang
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Binghui Xia
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Zhenghan Luo
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Zhenghao Xu
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China.
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Shanghai Key Laboratory of Medical Biodefense, Naval Medical University, Shanghai, China.
| |
Collapse
|
32
|
Salas-Rojas M, de Oliveira-Filho EF, Almazán-Marín C, Rodas-Martínez AZ, Aguilar-Setién Á, Drexler JF. Serological evidence for potential yellow fever virus infection in non-human primates, southeastern Mexico. ONE HEALTH OUTLOOK 2023; 5:14. [PMID: 37876014 PMCID: PMC10594671 DOI: 10.1186/s42522-023-00090-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Arthropod-borne flaviviruses like dengue virus (DENV) and yellow fever virus (YFV) are major human pathogens. In Latin America, YFV is maintained in sylvatic cycles involving non-human primates (NHP) and forest-dwelling mosquitos. YFV supposedly does not circulate north of Panama. METHODS We conducted a serologic study for flaviviruses and other emerging viruses in NHP from southeastern Mexico. A total of thirty sera of black-handed spider monkeys (Ateles geoffroyi, n = 25), black howler monkeys (Alouatta pigra, n = 3), and mantled howler monkeys (Al. palliata, n = 2) sampled in 2012 and 2018 were screened by an indirect immunofluorescence assay (IFA) to detected IgG antibodies against DENV, YFV, Zika virus (ZIKV), West Nile virus (WNV), Rift Valley fever virus, Crimean-Congo hemorrhagic fever virus, Middle East respiratory syndrome coronavirus, and Zaire Ebola virus, and confirmed by plaque reduction neutralization tests (PRNT90) representing all mosquito-borne flavivirus serocomplexes circulating in the Americas. RESULTS A total of 16 sera (53.3%; 95% CI, 34.3-71.7) showed IFA reactivity to at least one tested flavivirus with end-point titers ranging from 1:100 to 1:1000. No serum reacted with other viruses. Monotypic and high mean PRNT90 endpoint YFV titers of 1:246 were found in 3 black-handed spider monkey sera (10.0%; 95% CI, 2.1-26.5) sampled in 2018 in Tabasco, compared to all other flaviviruses tested. Monotypic endpoint PRNT90 titers of 1:28 for Ilheus virus and 1:22 for WNV in serum of black howler monkeys sampled in 2018 in Tabasco suggested additional flavivirus exposure. CONCLUSIONS Our findings may suggest unnoticed YFV circulation. Intensification of YFV surveillance in NHP and vectors is warranted in Mexico and potentially other areas considered free of yellow fever.
Collapse
Affiliation(s)
- Mónica Salas-Rojas
- UIM en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Edmilson Ferreira de Oliveira-Filho
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Cenia Almazán-Marín
- UIM en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Alba Zulema Rodas-Martínez
- División Académica de Ciencias Biológicas, Universidad Juárez Autónoma de Tabasco, Villahermosa, Tabasco, México
| | - Álvaro Aguilar-Setién
- UIM en Inmunología, UMAE Hospital de Pediatría, Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Jan Felix Drexler
- Institute of Virology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- German Centre for Infection Research (DZIF), Associated Partner Site Charité, Berlin, Germany.
| |
Collapse
|
33
|
Mesev EV, Lin AE, Guare EG, Heller BL, Douam F, Adamson B, Toettcher JE, Ploss A. Membrane-proximal motifs encode differences in signaling strength between type I and III interferon receptors. Sci Signal 2023; 16:eadf5494. [PMID: 37816090 PMCID: PMC10939449 DOI: 10.1126/scisignal.adf5494] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 09/09/2023] [Indexed: 10/12/2023]
Abstract
Interferons (IFNs) play crucial roles in antiviral defenses. Despite using the same Janus-activated kinase (JAK)-signal transducer and activator of transcription (STAT) signaling cascade, type I and III IFN receptors differ in the magnitude and dynamics of their signaling in terms of STAT phosphorylation, gene transcription, and antiviral responses. These differences are not due to ligand-binding affinity and receptor abundance. Here, we investigated the ability of the intracellular domains (ICDs) of IFN receptors to differentiate between type I and III IFN signaling. We engineered synthetic, heterodimeric type I and III IFN receptors that were stably expressed at similar amounts in human cells and responded to a common ligand. We found that our synthetic type I IFN receptors stimulated STAT phosphorylation and gene expression to greater extents than did the corresponding type III IFN receptors. Furthermore, we identified short "box motifs" within ICDs that bind to JAK1 that were sufficient to encode differences between the type I and III IFN receptors. Together, our results indicate that specific regions within the ICDs of IFN receptor subunits encode different downstream signaling strengths that enable type I and III IFN receptors to produce distinct signaling outcomes.
Collapse
Affiliation(s)
- Emily V. Mesev
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Aaron E. Lin
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Emma G. Guare
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Brigitte L. Heller
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Florian Douam
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA 02118, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA 02118, USA
| | - Britt Adamson
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Lewis Sigler Center for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Jared E. Toettcher
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
- Omenn-Darling Bioengineering Institute, Princeton University, Princeton, NJ 08544, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
34
|
Pardali V, Giannakopoulou E, Mpekoulis G, Tsopela V, Panos G, Taylor MC, Kelly JM, Vassilaki N, Zoidis G. Novel Lipophilic Hydroxamates Based on Spirocarbocyclic Hydantoin Scaffolds with Potent Antiviral and Trypanocidal Activity. Pharmaceuticals (Basel) 2023; 16:1046. [PMID: 37513957 PMCID: PMC10385743 DOI: 10.3390/ph16071046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Flaviviridae infections, such as those caused by hepatitis C (HCV) and dengue viruses (DENVs), represent global health risks. Infected people are in danger of developing chronic liver failure or hemorrhagic fever, both of which can be fatal if not treated. The tropical parasites Trypanosoma brucei and Trypanosoma cruzi cause enormous socioeconomic burdens in Sub-Saharan Africa and Latin America. Anti-HCV chemotherapy has severe adverse effects and is expensive, whereas dengue has no clinically authorized treatment. Antiparasitic medicines are often toxic and difficult to administer, and treatment failures are widely reported. There is an urgent need for new chemotherapies. Based on our previous research, we have undertaken structural modification of lead compound V with the goal of producing derivatives with both antiviral and trypanocidal activity. The novel spirocarbocyclic-substituted hydantoin analogs were designed, synthesized, and tested for antiviral activity against three HCV genotypes (1b, 3a, 4a), DENV, yellow fever virus (YFV), and two trypanosome species (T. brucei, T. cruzi). The optimization was successful and led to compounds with significant antiviral and trypanocidal activity and exceptional selectivity. Several modifications were made to further investigate the structure-activity relationships (SARs) and confirm the critical role of lipophilicity and conformational degrees of freedom.
Collapse
Affiliation(s)
- Vasiliki Pardali
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - Erofili Giannakopoulou
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| | - George Mpekoulis
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Vassilina Tsopela
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Georgios Panos
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Martin C Taylor
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - John M Kelly
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Niki Vassilaki
- Molecular Virology Laboratory, Hellenic Pasteur Institute, Vas. Sofias Avenue, 11521 Athens, Greece
| | - Grigoris Zoidis
- School of Health Sciences, Department of Pharmacy, Division of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zografou, 15771 Athens, Greece
| |
Collapse
|
35
|
Silva NIO, Albery GF, Arruda MS, Oliveira GFG, Costa TA, de Mello ÉM, Moreira GD, Reis EV, da Silva SA, Silva MC, de Almeida MG, Becker DJ, Carlson CJ, Vasilakis N, Hanley KA, Drumond BP. Ecological drivers of sustained enzootic yellow fever virus transmission in Brazil, 2017-2021. PLoS Negl Trop Dis 2023; 17:e0011407. [PMID: 37276217 PMCID: PMC10270639 DOI: 10.1371/journal.pntd.0011407] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 06/15/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
Beginning December 2016, sylvatic yellow fever (YF) outbreaks spread into southeastern Brazil, and Minas Gerais state experienced two sylvatic YF waves (2017 and 2018). Following these massive YF waves, we screened 187 free-living non-human primate (NHPs) carcasses collected throughout the state between January 2019 and June 2021 for YF virus (YFV) using RTqPCR. One sample belonging to a Callithrix, collected in June 2020, was positive for YFV. The viral strain belonged to the same lineage associated with 2017-2018 outbreaks, showing the continued enzootic circulation of YFV in the state. Next, using data from 781 NHPs carcasses collected in 2017-18, we used generalized additive mixed models (GAMMs) to identify the spatiotemporal and host-level drivers of YFV infection and intensity (an estimation of genomic viral load in the liver of infected NHP). Our GAMMs explained 65% and 68% of variation in virus infection and intensity, respectively, and uncovered strong temporal and spatial patterns for YFV infection and intensity. NHP infection was higher in the eastern part of Minas Gerais state, where 2017-2018 outbreaks affecting humans and NHPs were concentrated. The odds of YFV infection were significantly lower in NHPs from urban areas than from urban-rural or rural areas, while infection intensity was significantly lower in NHPs from urban areas or the urban-rural interface relative to rural areas. Both YFV infection and intensity were higher during the warm/rainy season compared to the cold/dry season. The higher YFV intensity in NHPs in warm/rainy periods could be a result of higher exposure to vectors and/or higher virus titers in vectors during this time resulting in the delivery of a higher virus dose and higher viral replication levels within NHPs. Further studies are needed to better test this hypothesis and further compare the dynamics of YFV enzootic cycles between different seasons.
Collapse
Affiliation(s)
| | - Gregory F. Albery
- Department of Biology, Georgetown University, Washington, DC, United States of America
| | - Matheus Soares Arruda
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Thaís Alkifeles Costa
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Érica Munhoz de Mello
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Gabriel Dias Moreira
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Erik Vinícius Reis
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone Agostinho da Silva
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Marlise Costa Silva
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Munique Guimarães de Almeida
- Laboratório de Zoonoses—Centro de Controle de Zoonoses, Prefeitura de Belo Horizonte, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel J. Becker
- Department of Biology, University of Oklahoma, Norman, Oklahoma, United States of America
| | - Colin J. Carlson
- Department of Biology, Georgetown University, Washington, DC, United States of America
- Center for Global Health Science and Security, Georgetown University, Washington, D.C., United States of America
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Vector-Borne and Zoonotic Diseases, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kathryn A. Hanley
- Department of Biology, New Mexico State University, Las Cruces, New Mexico, United States of America
| | - Betânia Paiva Drumond
- Department of Microbiology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
36
|
Hale GL. Flaviviruses and the Traveler: Around the World and to Your Stage. A Review of West Nile, Yellow Fever, Dengue, and Zika Viruses for the Practicing Pathologist. Mod Pathol 2023; 36:100188. [PMID: 37059228 DOI: 10.1016/j.modpat.2023.100188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023]
Abstract
Flaviviruses are a genus of single-stranded RNA viruses that impose an important and growing burden to human health. There are over 3 billion individuals living in areas where flaviviruses are endemic. Flaviviruses and their arthropod vectors (which include mosquitoes and ticks) take advantage of global travel to expand their distribution and cause severe disease in humans, and they can be grouped according to their vector and pathogenicity. The mosquito-borne flaviviruses cause a spectrum of diseases from encephalitis to hepatitis and vascular shock syndrome, congenital abnormalities, and fetal death. Neurotropic infections such as Zika virus and West Nile virus cross the blood-brain barrier and infect neurons and other cells, leading to meningoencephalitis. In the hemorrhagic fever clade, there are yellow fever virus, the prototypical hemorrhagic fever virus that infects hepatocytes, and dengue virus, which infects cells of the reticuloendothelial system and can lead to a dramatic plasma cell leakage and shock syndrome. Zika virus also causes congenital infections and fetal death and is the first and only example of a teratogenic arbovirus in humans. Diagnostic testing for flaviviruses broadly includes the detection of viral RNA in serum (particularly within the first 10 days of symptoms), viral isolation by cell culture (rarely performed due to complexity and biosafety concerns), and histopathologic evaluation with immunohistochemistry and molecular testing on formalin-fixed paraffin-embedded tissue blocks. This review focuses on 4 mosquito-borne flaviviruses-West Nile, yellow fever, dengue, and Zika virus-and discusses the mechanisms of transmission, the role of travel in geographic distribution and epidemic emergence, and the clinical and histopathologic features of each. Finally, prevention strategies such as vector control and vaccination are discussed.
Collapse
Affiliation(s)
- Gillian L Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
37
|
Bhattacharjee S, Ghosh D, Saha R, Sarkar R, Kumar S, Khokhar M, Pandey RK. Mechanism of Immune Evasion in Mosquito-Borne Diseases. Pathogens 2023; 12:635. [PMID: 37242305 PMCID: PMC10222277 DOI: 10.3390/pathogens12050635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, mosquito-borne illnesses have emerged as a major health burden in many tropical regions. These diseases, such as malaria, dengue fever, chikungunya, yellow fever, Zika virus infection, Rift Valley fever, Japanese encephalitis, and West Nile virus infection, are transmitted through the bite of infected mosquitoes. These pathogens have been shown to interfere with the host's immune system through adaptive and innate immune mechanisms, as well as the human circulatory system. Crucial immune checkpoints such as antigen presentation, T cell activation, differentiation, and proinflammatory response play a vital role in the host cell's response to pathogenic infection. Furthermore, these immune evasions have the potential to stimulate the human immune system, resulting in other associated non-communicable diseases. This review aims to advance our understanding of mosquito-borne diseases and the immune evasion mechanisms by associated pathogens. Moreover, it highlights the adverse outcomes of mosquito-borne disease.
Collapse
Affiliation(s)
| | - Debanjan Ghosh
- Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Rounak Saha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Rima Sarkar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Saurav Kumar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS, Jodhpur 342005, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Sweden
| |
Collapse
|
38
|
Fradico JRB, Campi-Azevedo AC, Speziali E, do Valle Antonelli LR, Peruhype-Magalhães V, de Rezende IM, Alves PA, Pascoal-Xavier MA, Pereira LS, Dutra MRT, Ramalho DB, Cenachi A, de Paula L, Santos TA, do Carmo Said RF, Calzavara-Silva CE, Coelho-Dos-Reis JGA, de Magalhães CR, Rabelo LLC, Valim V, Brito-de-Sousa JP, da Costa-Rocha IA, de Souza Gomes M, Amaral LR, de Lima SMB, Trindade GF, Santos RT, da Silva JFA, Monath T, LaBeaud AD, Drumond BP, Martins-Filho OA, Teixeira-Carvalho A. Serum soluble mediators as prognostic biomarkers for morbidity, disease outcome, and late-relapsing hepatitis in yellow fever patients. Clin Immunol 2023; 251:109321. [PMID: 37019421 DOI: 10.1016/j.clim.2023.109321] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
This study described a soluble mediator storm in acute Yellow Fever/YF infection along the kinetic timeline towards convalescent disease. The analyses of the YF Viral RNAnemia, chemokines, cytokines, and growth factors were performed in YF patients at acute/(D1-15) and convalescent/(D16-315) phases. Patients with acute YF infection displayed a trimodal viremia profile spreading along D3, D6, and D8-14. A massive storm of mediators was observed in acute YF. Higher levels of mediators were observed in YF with higher morbidity scores, patients under intensive care, and those progressing to death than in YF patients who progress to late-relapsing hepatitis/L-Hep. A unimodal peak of biomarkers around D4-6 with a progressive decrease towards D181-315 was observed in non-L-Hep patients, while a bimodal pattern with a second peak around D61-90 was associated with L-Hep. This study provided a comprehensive landscape of evidence that distinct immune responses drive pathogenesis, disease progression, and L-Hep in YF patients.
Collapse
Affiliation(s)
| | | | - Elaine Speziali
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | | | | | - Izabela Maurício de Rezende
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pedro Augusto Alves
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil
| | - Marcelo Antônio Pascoal-Xavier
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil; Departamento de Anatomia Patológica e Medicina Legal, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Leonardo Soares Pereira
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | - Maria Rita Teixeira Dutra
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | - Dario Brock Ramalho
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | - Adriana Cenachi
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | - Ludmila de Paula
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | - Tayrine Araujo Santos
- Hospital Eduardo de Menezes (HEM), Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, MG, Brazil
| | | | | | - Jordana Grazziela Alves Coelho-Dos-Reis
- Instituto René Rachou, Fundação Oswaldo Cruz (FIOCRUZ-Minas), Belo Horizonte, MG, Brazil; Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Valéria Valim
- Hospital Universitário Cassiano Antônio Moraes, Universidade Federal do Espírito Santo (HUCAM/UFES/EBSERH), Vitória, ES, Brazil
| | | | | | - Matheus de Souza Gomes
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, MG, Brazil
| | - Laurence Rodrigues Amaral
- Laboratório de Bioinformática e Análises Moleculares, Universidade Federal de Uberlândia, Campus Patos de Minas, MG, Brazil
| | - Sheila Maria Barbosa de Lima
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Gisela Freitas Trindade
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Renata Tourinho Santos
- Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | | | | | - Angelle Desiree LaBeaud
- Department of Pediatrics, Infectious Disease Division, Stanford University School of Medicine, Stanford, CA, USA
| | - Betânia Paiva Drumond
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | |
Collapse
|
39
|
Dutta SK, Langenburg T. A Perspective on Current Flavivirus Vaccine Development: A Brief Review. Viruses 2023; 15:v15040860. [PMID: 37112840 PMCID: PMC10142581 DOI: 10.3390/v15040860] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/09/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
The flavivirus genus contains several clinically important pathogens that account for tremendous global suffering. Primarily transmitted by mosquitos or ticks, these viruses can cause severe and potentially fatal diseases ranging from hemorrhagic fevers to encephalitis. The extensive global burden is predominantly caused by six flaviviruses: dengue, Zika, West Nile, yellow fever, Japanese encephalitis and tick-borne encephalitis. Several vaccines have been developed, and many more are currently being tested in clinical trials. However, flavivirus vaccine development is still confronted with many shortcomings and challenges. With the use of the existing literature, we have studied these hurdles as well as the signs of progress made in flavivirus vaccinology in the context of future development strategies. Moreover, all current licensed and phase-trial flavivirus vaccines have been gathered and discussed based on their vaccine type. Furthermore, potentially relevant vaccine types without any candidates in clinical testing are explored in this review as well. Over the past decades, several modern vaccine types have expanded the field of vaccinology, potentially providing alternative solutions for flavivirus vaccines. These vaccine types offer different development strategies as opposed to traditional vaccines. The included vaccine types were live-attenuated, inactivated, subunit, VLPs, viral vector-based, epitope-based, DNA and mRNA vaccines. Each vaccine type offers different advantages, some more suitable for flaviviruses than others. Additional studies are needed to overcome the barriers currently faced by flavivirus vaccine development, but many potential solutions are currently being explored.
Collapse
|
40
|
Wu B, Qi Z, Qian X. Recent Advancements in Mosquito-Borne Flavivirus Vaccine Development. Viruses 2023; 15:813. [PMID: 37112794 PMCID: PMC10143207 DOI: 10.3390/v15040813] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lately, the global incidence of flavivirus infection has been increasing dramatically and presents formidable challenges for public health systems around the world. Most clinically significant flaviviruses are mosquito-borne, such as the four serotypes of dengue virus, Zika virus, West Nile virus, Japanese encephalitis virus and yellow fever virus. Until now, no effective antiflaviviral drugs are available to fight flaviviral infection; thus, a highly immunogenic vaccine would be the most effective weapon to control the diseases. In recent years, flavivirus vaccine research has made major breakthroughs with several vaccine candidates showing encouraging results in preclinical and clinical trials. This review summarizes the current advancement, safety, efficacy, advantages and disadvantages of vaccines against mosquito-borne flaviviruses posing significant threats to human health.
Collapse
Affiliation(s)
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| | - Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai 200433, China;
| |
Collapse
|
41
|
Sandberg JT, Löfling M, Varnaitė R, Emgård J, Al-Tawil N, Lindquist L, Gredmark-Russ S, Klingström J, Loré K, Blom K, Ljunggren HG. Safety and immunogenicity following co-administration of Yellow fever vaccine with Tick-borne encephalitis or Japanese encephalitis vaccines: Results from an open label, non-randomized clinical trial. PLoS Negl Trop Dis 2023; 17:e0010616. [PMID: 36758067 PMCID: PMC9946270 DOI: 10.1371/journal.pntd.0010616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 02/22/2023] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Flavivirus infections pose a significant global health burden underscoring the need for the development of safe and effective vaccination strategies. Available flavivirus vaccines are from time to time concomitantly delivered to individuals. Co-administration of different vaccines saves time and visits to health care units and vaccine clinics. It serves to provide protection against multiple pathogens in a shorter time-span; e.g., for individuals travelling to different endemic areas. However, safety and immunogenicity-related responses have not been appropriately evaluated upon concomitant delivery of these vaccines. Therefore, we performed an open label, non-randomized clinical trial studying the safety and immunogenicity following concomitant delivery of the yellow fever virus (YFV) vaccine with tick-borne encephalitis virus (TBEV) and Japanese encephalitis virus (JE) virus vaccines. METHODS AND FINDINGS Following screening, healthy study participants were enrolled into different cohorts receiving either TBEV and YFV vaccines, JEV and YFV vaccines, or in control groups receiving only the TBEV, JEV, or YFV vaccine. Concomitant delivery was given in the same or different upper arms for comparison in the co-vaccination cohorts. Adverse effects were recorded throughout the study period and blood samples were taken before and at multiple time-points following vaccination to evaluate immunological responses to the vaccines. Adverse events were predominantly mild in the study groups. Four serious adverse events (SAE) were reported, none of them deemed related to vaccination. The development of neutralizing antibodies (nAbs) against TBEV, JEV, or YFV was not affected by the concomitant vaccination strategy. Concomitant vaccination in the same or different upper arms did not significantly affect safety or immunogenicity-related outcomes. Exploratory studies on immunological effects were additionally performed and included studies of lymphocyte activation, correlates associated with germinal center activation, and plasmablast expansion. CONCLUSIONS Inactivated TBEV or JEV vaccines can be co-administered with the live attenuated YFV vaccine without an increased risk of adverse events and without reduced development of nAbs to the respective viruses. The vaccines can be delivered in the same upper arm without negative outcome. In a broader perspective, the results add valuable information for simultaneous administration of live and inactivated flavivirus vaccines in general. TRIAL REGISTRATION Eudra CT 2017-002137-32.
Collapse
Affiliation(s)
- John Tyler Sandberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marie Löfling
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Renata Varnaitė
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Emgård
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Nabil Al-Tawil
- Karolinska Trial Alliance, Karolinska University Hospital, Stockholm, Sweden
| | - Lars Lindquist
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Gredmark-Russ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Kim Blom
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
42
|
Howard-Jones AR, Pham D, Sparks R, Maddocks S, Dwyer DE, Kok J, Basile K. Arthropod-Borne Flaviviruses in Pregnancy. Microorganisms 2023; 11:433. [PMID: 36838398 PMCID: PMC9959669 DOI: 10.3390/microorganisms11020433] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Flaviviruses are a diverse group of enveloped RNA viruses that cause significant clinical manifestations in the pregnancy and postpartum periods. This review highlights the epidemiology, pathophysiology, clinical features, diagnosis, and prevention of the key arthropod-borne flaviviruses of concern in pregnancy and the neonatal period-Zika, Dengue, Japanese encephalitis, West Nile, and Yellow fever viruses. Increased disease severity during pregnancy, risk of congenital malformations, and manifestations of postnatal infection vary widely amongst this virus family and may be quite marked. Laboratory confirmation of infection is complex, especially due to the reliance on serology for which flavivirus cross-reactivity challenges diagnostic specificity. As such, a thorough clinical history including relevant geographic exposures and prior vaccinations is paramount for accurate diagnosis. Novel vaccines are eagerly anticipated to ameliorate the impact of these flaviviruses, particularly neuroinvasive disease manifestations and congenital infection, with consideration of vaccine safety in pregnant women and children pivotal. Moving forward, the geographical spread of flaviviruses, as for other zoonoses, will be heavily influenced by climate change due to the potential expansion of vector and reservoir host habitats. Ongoing 'One Health' engagement across the human-animal-environment interface is critical to detect and responding to emergent flavivirus epidemics.
Collapse
Affiliation(s)
- Annaleise R. Howard-Jones
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
| | - David Pham
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Rebecca Sparks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Susan Maddocks
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
| | - Dominic E. Dwyer
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Infectious Diseases and Microbiology-Public Health, Westmead, NSW 2145, Australia
| | - Jen Kok
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
- Centre for Infectious Diseases and Microbiology-Public Health, Westmead, NSW 2145, Australia
| | - Kerri Basile
- Centre for Infectious Diseases and Microbiology Laboratory Services, Institute of Clinical Pathology and Medical Research—NSW Health Pathology, Westmead, NSW 2145, Australia
- Sydney Infectious Diseases Institute, The University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
43
|
Potential Mammalian Vector-Borne Diseases in Live and Wet Markets in Indonesia and Myanmar. MICROBIOLOGY RESEARCH 2023. [DOI: 10.3390/microbiolres14010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Vector-borne diseases spread from wild animals and their associated ectoparasites to humans and domesticated animals. Wildlife markets are recognized as important areas where this transfer can take place. We assessed the potential for spreading vector-borne diseases in two live and wet markets in Myanmar (Mong La, on the Myanmar-China border) and Indonesia (Sukahaji in Bandung on the island of Java) by making an inventory of all live and freshly killed wild mammals for sale. For eight mammal families, we quantified the number of animals on offer, and we used a heatmap cluster analysis to map vector-borne diseases that these families may carry. In Myanmar, we observed large numbers of wild pigs and deer (potentially carrying West Nile and various encephalitis viruses) whereas in Indonesia we observed Old World fruit bats (potentially carrying Chikungunya and encephalitis viruses) and squirrels (potentially carrying West Nile and encephalitis viruses). The trade in Indonesia was dominated by live mammals offered for sale as pets, and only Old World fruit bats and squirrels traded for traditional Asian medicine were killed in the markets. The trade in Myanmar was more geared towards wild meat (e.g., wild pigs, deer, primates) and traditional Asian medicine (squirrels). The combined risks of vector-borne diseases spreading from traded animals to human health highlight the need for an integrated approach protecting public health, economic interests and biodiversity.
Collapse
|
44
|
Montalvo Zurbia-Flores G, Rollier CS, Reyes-Sandoval A. Re-thinking yellow fever vaccines: fighting old foes with new generation vaccines. Hum Vaccin Immunother 2022; 18:1895644. [PMID: 33974507 PMCID: PMC8920179 DOI: 10.1080/21645515.2021.1895644] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/07/2021] [Accepted: 02/21/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the existence of a highly efficient yellow fever vaccine, yellow fever reemergence throughout Africa and the Americas has put 900 million people in 47 countries at risk of contracting the disease. Although the vaccine has been key to controlling yellow fever epidemics, its live-attenuated nature comes with a range of contraindications that prompts advising against its administration to pregnant and lactating women, immunocompromised individuals, and those with hypersensitivity to chicken egg proteins. Additionally, large outbreaks have highlighted problems with insufficient vaccine supply, whereby manufacturers rely on slow traditional manufacturing processes that prevent them from ramping up production. These limitations have contributed to an inadequate control of yellow fever and have favored the pursuit of novel yellow fever vaccine candidates that aim to circumvent the licensed vaccine's restrictions. Here, we review the live-attenuated vaccine's limitations and explore the epitome of a yellow fever vaccine, whilst scrutinizing next-generation vaccine candidates.
Collapse
Affiliation(s)
- Gerardo Montalvo Zurbia-Flores
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
| | - Christine S Rollier
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Arturo Reyes-Sandoval
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford. The Henry Wellcome Building for Molecular Physiology, Oxford, UK
- Instituto Politécnico Nacional, IPN. Av. Luis Enrique Erro S/n. Unidad Adolfo López Mateos. CP, Mexico City, Mexico
| |
Collapse
|
45
|
Ong J, Ho SH, Soh SXH, Wong Y, Ng Y, Vasquez K, Lai YL, Setoh YX, Chong CS, Lee V, Wong JCC, Tan CH, Sim S, Ng LC, Lim JT. Assessing the efficacy of male Wolbachia-infected mosquito deployments to reduce dengue incidence in Singapore: study protocol for a cluster-randomized controlled trial. Trials 2022; 23:1023. [PMID: 36528590 PMCID: PMC9758775 DOI: 10.1186/s13063-022-06976-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Dengue is a severe environmental public health challenge in tropical and subtropical regions. In Singapore, decreasing seroprevalence and herd immunity due to successful vector control has paradoxically led to increased transmission potential of the dengue virus. We have previously demonstrated that incompatible insect technique coupled with sterile insect technique (IIT-SIT), which involves the release of X-ray-irradiated male Wolbachia-infected mosquitoes, reduced the Aedes aegypti population by 98% and dengue incidence by 88%. This novel vector control tool is expected to be able to complement current vector control to mitigate the increasing threat of dengue on a larger scale. We propose a multi-site protocol to study the efficacy of IIT-SIT at reducing dengue incidence. METHODS/DESIGN The study is designed as a parallel, two-arm, non-blinded cluster-randomized (CR) controlled trial to be conducted in high-rise public housing estates in Singapore, an equatorial city-state. The aim is to determine whether large-scale deployment of male Wolbachia-infected Ae. aegypti mosquitoes can significantly reduce dengue incidence in intervention clusters. We will use the CR design, with the study area comprising 15 clusters with a total area of 10.9 km2, covering approximately 722,204 residents in 1713 apartment blocks. Eight clusters will be randomly selected to receive the intervention, while the other seven will serve as non-intervention clusters. Intervention efficacy will be estimated through two primary endpoints: (1) odds ratio of Wolbachia exposure distribution (i.e., probability of living in an intervention cluster) among laboratory-confirmed reported dengue cases compared to test-negative controls and (2) laboratory-confirmed reported dengue counts normalized by population size in intervention versus non-intervention clusters. DISCUSSION This study will provide evidence from a multi-site, randomized controlled trial for the efficacy of IIT-SIT in reducing dengue incidence. The trial will provide valuable information to estimate intervention efficacy for this novel vector control approach and guide plans for integration into national vector control programs in dengue-endemic settings. TRIAL REGISTRATION ClinicalTrials.gov, identifier: NCT05505682 . Registered on 16 August 2022. Retrospectively registered.
Collapse
Affiliation(s)
- Janet Ong
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Soon Hoe Ho
- Environmental Health Institute, National Environment Agency, Singapore, Singapore.
| | - Stacy Xin Hui Soh
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Yvonne Wong
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Youming Ng
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Kathryn Vasquez
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Yee Ling Lai
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Yin Xiang Setoh
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Chee-Seng Chong
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Vernon Lee
- Communicable Diseases Division, Ministry of Health, Singapore, Singapore
| | | | - Cheong Huat Tan
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Shuzhen Sim
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Lee Ching Ng
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
| | - Jue Tao Lim
- Environmental Health Institute, National Environment Agency, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Novena Campus, Singapore, Singapore
| |
Collapse
|
46
|
Saito K, Shimasaki K, Fukasawa M, Suzuki R, Okemoto-Nakamura Y, Katoh K, Takasaki T, Hanada K. Establishment of Vero cell lines persistently harboring a yellow fever virus 17D subgenomic replicon. Virus Res 2022; 322:198935. [PMID: 36152929 DOI: 10.1016/j.virusres.2022.198935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022]
Abstract
Yellow fever virus (YFV), a member of the genus Flavivirus, family Flaviviridae, is the etiological agent for an acute viral hemorrhagic disease, yellow fever. Although effective live attenuated vaccines based on the strain YFV 17D are currently available, no specific antiviral drug is available, and the disease remains a major public health concern. Hence, the discovery and development of antiviral drugs should lead to great benefits in controlling the disease. To provide a screening platform for antiviral agents targeting YFV RNA translation/replication, we have established and characterized two Vero cell lines that persistently harbor a subgenomic replicon derived from YFV 17D-204 (referred to as replicon cells). The replicon carries YFV nucleotides (1 - 176 and 2382-10,862) and a green fluorescent protein (GFP)-Zeocin resistance fusion gene as a selection marker and indicator of persistent replication. Immunofluorescence analysis revealed that both replicon cells and YFV 17D-infected cells showed similar distribution patterns of viral NS4B protein and replication intermediate, double-stranded RNA. Sequencing analysis of persistent replicons from the two replicon cell lines suggested that their nucleotide sequences did not vary greatly following multiple passages. We examined the effect of five agents, the antiviral cytokines interferon-β and -γ, the nucleoside analog ribavirin, the squalene synthase inhibitor zaragozic acid A, and the antibiotic rifapentine, a recently reported entry and replication inhibitor against YFV, on the persistent replication in the two replicon cell lines. These agents were selected because they inhibited both production of YFV 17D and transient replication of a luciferase-expressing replicon in Vero cells, without greatly affecting cell viability. We found that each of the agents decreased GFP fluorescence in the replicon cells, albeit to varying degrees. The agents other than rifapentine also showed a decrease in viral RNA levels in the replicon cells comparable to that seen for GFP fluorescence. These results indicate that persistent replication is susceptible to each of these five agents, although their mechanisms of action may differ. Taken together, these results provide evidence that translation/replication of the replicon in the replicon cells mimics that of the viral genome upon YFV 17D infection, indicating that the replicon cell lines can serve as a useful tool for high-throughput antiviral drug screening.
Collapse
Affiliation(s)
- Kyoko Saito
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan.
| | - Kentaro Shimasaki
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Masayoshi Fukasawa
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Ryosuke Suzuki
- Department of Virology II, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, Japan
| | - Yuko Okemoto-Nakamura
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Kaoru Katoh
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba-shi, Ibaragi, Japan; AIRC, National Institute of Advanced Industrial Science and Technology (AIST), Koto-ku, Tokyo, Japan
| | - Tomohiko Takasaki
- Kanagawa Prefectural Institute of Public Health, Chigasaki-shi, Kanagawa, Japan
| | - Kentaro Hanada
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan; Department of Quality Assurance, Radiation Safety, and Information System, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
47
|
Mohapatra RK, Kutikuppala SLV, Ansari A, Kandi V, Mishra S. Another neglected tropical disease yellow fever re-emerges in African countries: Potential threat in the COVID-19 era which needs comprehensive investigations - Correspondence. Int J Surg 2022; 108:106988. [PMID: 36368420 PMCID: PMC9643270 DOI: 10.1016/j.ijsu.2022.106988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, 758 002, Odisha, India Department of General Surgery, Dr NTR University of Health Sciences, Vijayawada, Andhra Pradesh, India Department of Chemistry, Central University of Haryana, Mahendergarh, Haryana, 123031, India Department of Microbiology, Prathima Institute of Medical Sciences, Karimnagar, 505417, Telangana, India School of Biotechnology, Campus-11, KIIT Deemed-to-be-University, Bhubaneswar, Odisha, 751 024, India
| | | | | | | | | |
Collapse
|
48
|
Bailey AL, Diamond MS. Hepatopathology of flaviviruses. J Hepatol 2022; 77:1711-1713. [PMID: 35981935 DOI: 10.1016/j.jhep.2022.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 12/04/2022]
Affiliation(s)
- Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Heath, University of Wisconsin-Madison, Madison, WI, USA.
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
49
|
Jung L, Schneider A, Lübbert C. Gelbfieber. ZEITSCHRIFT FÜR GASTROENTEROLOGIE 2022. [DOI: 10.1055/a-1965-4267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Qian X, Wu B, Tang H, Luo Z, Xu Z, Ouyang S, Li X, Xie J, Yi Z, Leng Q, Liu Y, Qi Z, Zhao P. Rifapentine is an entry and replication inhibitor against yellow fever virus both in vitro and in vivo. Emerg Microbes Infect 2022; 11:873-884. [PMID: 35249454 PMCID: PMC8942558 DOI: 10.1080/22221751.2022.2049983] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Yellow fever virus (YFV) infection is a major public concern that threatens a large population in South America and Africa. No specific anti-YFV drugs are available till now. Here, we report that rifapentine is a potent YFV inhibitor in various cell lines by high-throughput drugs screening, acting at both cell entry and replication steps. Kinetic test and binding assay suggest that rifapentine interferes the viral attachment to the target cells. The application of YFV replicon and surface plasmon resonance assay indicates that rifapentine suppresses viral replication by binding to the RNA-dependent RNA polymerase (RdRp) domain of viral nonstructural protein NS5. Further molecular docking suggests that it might interact with the active centre of RdRp. Rifapentine significantly improves the survival rate, alleviates clinical signs, and reduces virus load and injury in targeted organs both in YFV-infected type I interferon receptor knockout A129−/− and wild-type C57 mice. The antiviral effect in vivo is robust during both prophylactic intervention and therapeutic treatment, and the activity is superior to sofosbuvir, a previously reported YFV inhibitor in mice. Our data show that rifapentine may serve as an effective anti-YFV agent, providing promising prospects in the development of YFV pharmacotherapy.
Collapse
Affiliation(s)
- Xijing Qian
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Bingan Wu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Hailin Tang
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Zhenghan Luo
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Zhenghao Xu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Songying Ouyang
- Key Laboratory of Innate Immune Biology of Fujian Province, College of Life Sciences, Fujian Normal University, Fujian, People's Republic of China
| | - Xiangliang Li
- Key Laboratory of Innate Immune Biology of Fujian Province, College of Life Sciences, Fujian Normal University, Fujian, People's Republic of China
| | - Jianfeng Xie
- Fujian Provincial Center for Disease Control and Prevention, Fujian, People's Republic of China
| | - Zhigang Yi
- Key Laboratory of Medical Molecular Virology and Department of Medical Microbiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Qibin Leng
- State Key Laboratory of Respiratory Diseases, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yan Liu
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Zhongtian Qi
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| | - Ping Zhao
- Department of Microbiology, Faculty of Naval Medicine, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|