1
|
Chen GH, Dai YC, Hsieh SC, Tsai JJ, Sy AK, Jiz M, Pedroso C, Brites C, Netto EM, Kanki PJ, Saunders DRD, Vanlandingham DL, Higgs S, Huang YJS, Wang WK. Detection of anti-premembrane antibody as a specific marker of four flavivirus serocomplexes and its application to serosurveillance in endemic regions. Emerg Microbes Infect 2024; 13:2301666. [PMID: 38163752 PMCID: PMC10810658 DOI: 10.1080/22221751.2023.2301666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/30/2023] [Indexed: 01/03/2024]
Abstract
In the past few decades, several emerging/re-emerging mosquito-borne flaviviruses have resulted in disease outbreaks of public health concern in the tropics and subtropics. Due to cross-reactivities of antibodies recognizing the envelope protein of different flaviviruses, serosurveillance remains a challenge. Previously we reported that anti-premembrane (prM) antibody can discriminate between three flavivirus infections by Western blot analysis. In this study, we aimed to develop a serological assay that can discriminate infection or exposure with flaviviruses from four serocomplexes, including dengue (DENV), Zika (ZIKV), West Nile (WNV) and yellow fever (YFV) viruses, and explore its application for serosurveillance in flavivirus-endemic countries. We employed Western blot analysis including antigens of six flaviviruses (DENV1, 2 and 4, WNV, ZIKV and YFV) from four serocomplexes. We tested serum samples from YF-17D vaccinees, and from DENV, ZIKV and WNV panels that had been confirmed by RT-PCR or by neutralization assays. The overall sensitivity/specificity of anti-prM antibodies for DENV, ZIKV, WNV, and YFV infections/exposure were 91.7%/96.4%, 91.7%/99.2%, 88.9%/98.3%, and 91.3%/92.5%, respectively. When testing 48 samples from Brazil, we identified multiple flavivirus infections/exposure including DENV and ZIKV, DENV and YFV, and DENV, ZIKV and YFV. When testing 50 samples from the Philippines, we detected DENV, ZIKV, and DENV and ZIKV infections with a ZIKV seroprevalence rate of 10%, which was consistent with reports of low-level circulation of ZIKV in Asia. Together, these findings suggest that anti-prM antibody is a flavivirus serocomplex-specific marker and can be employed to delineate four flavivirus infections/exposure in regions where multiple flaviviruses co-circulate.
Collapse
Affiliation(s)
- Guan-Hua Chen
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Yu-Ching Dai
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Szu-Chia Hsieh
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jih-Jin Tsai
- Tropical Medicine Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Division of Infectious Diseases, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ava Kristy Sy
- National Reference Laboratory for Dengue and Other Arbovirus, Virology Department, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Mario Jiz
- Immunology Department, Research Institute for Tropical Medicine, Muntinlupa City, Philippines
| | - Celia Pedroso
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Carlos Brites
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Eduardo Martins Netto
- LAPI-Laboratório de Pesquisa em Infectologia-School of Medicine, Federal University of Bahia, Salvador, Brazil
| | - Phyllis J. Kanki
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Dana L. Vanlandingham
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Stephen Higgs
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Yan-Jang S. Huang
- Biosecurity Research Institute and Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Wei-Kung Wang
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
2
|
Vieira CJSP, Gyawali N, Onn MB, Shivas MA, Shearman D, Darbro JM, Wallau GL, van den Hurk AF, Frentiu FD, Skinner EB, Devine GJ. Mosquito bloodmeals can be used to determine vertebrate diversity, host preference, and pathogen exposure in humans and wildlife. Sci Rep 2024; 14:23203. [PMID: 39369026 PMCID: PMC11455984 DOI: 10.1038/s41598-024-73820-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
The surveillance and detection of zoonotic pathogens in animals is essential for predicting disease transmission pathways and the risks of spillover, but challenges include the costs, ethics and technical expertise required for vertebrate trapping, serum sampling and antibody or virus screening. Surveillance using haematophagous arthropods as a sampling tool offers a unique opportunity to obtain blood samples from a wide range of vertebrate species, allowing the study of host-mosquito associations, and host exposure to pathogens. We explored vertebrate diversity and potential Ross River virus (RRV) transmission pathways by analysing blood-fed mosquitoes collected in Brisbane, Australia. Host origins were identified using barcode sequencing, and host exposure to RRV was assessed using a modified plaque reduction neutralisation test. In total, 480 blood-fed mosquitoes were collected between February 2021 and May 2022. The host origins of 346 (72%) bloodmeals were identified, with humans (73%) and cattle (9%) comprising the dominant hosts. RRV seroprevalence was high in both vertebrate species with evidence of RRV exposure in 70% (21/30) of cattle and 52% (132/253) of humans. This is a novel, non-invasive method of estimating seroprevalence in vertebrate host populations. Our results highlight the potential of blood-fed mosquitoes to provide species-specific insights into pathogen transmission dynamics.
Collapse
Affiliation(s)
- Carla Julia S P Vieira
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia.
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4006, Australia.
| | - Narayan Gyawali
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Michael B Onn
- Entomology Laboratory, Public Space Operations, Brisbane City Council, Brisbane, QLD, 4009, Australia
| | - Martin A Shivas
- Entomology Laboratory, Public Space Operations, Brisbane City Council, Brisbane, QLD, 4009, Australia
| | - Damien Shearman
- Metro North Public Health Unit, Queensland Health, Brisbane, QLD, 4030, Australia
| | - Jonathan M Darbro
- Metro North Public Health Unit, Queensland Health, Brisbane, QLD, 4030, Australia
| | - Gabriel L Wallau
- Department of Entomology and Bioinformatic Core of the Aggeu Magalhães Institute, Oswaldo Cruz Foundation, Recife, 50740-465, PE, Brazil
- Department of Arbovirology, Bernhard Nocht Institute for Tropical Medicine, WHO Collaborating Center for Arbovirus and Hemorrhagic Fever Reference and Research, National Reference Center for Tropical Infectious Diseases, 20359, Hamburg, Germany
| | - Andrew F van den Hurk
- Public Health Virology, Forensic and Scientific Services, Queensland Health, Brisbane, QLD, 4108, Australia
| | - Francesca D Frentiu
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4006, Australia
| | - Eloise B Skinner
- Department of Biology, Stanford University, Stanford, CA, 94305, USA
- Centre for Planetary Health and Food Security, Griffith University, Gold Coast, QLD, 4215, Australia
| | - Gregor J Devine
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| |
Collapse
|
3
|
Sharma A, Chandrashekar CR, Krishna S, Sowdhamini R. Computational Analysis of the Accumulation of Mutations in Therapeutically Important RNA Viral Proteins During Pandemics with Special Emphasis on SARS-CoV-2. J Mol Biol 2024; 436:168716. [PMID: 39047897 DOI: 10.1016/j.jmb.2024.168716] [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: 04/07/2024] [Revised: 07/06/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Single stranded RNA viruses are primary causative agents for pandemics, causing extensive morbidity and mortality worldwide. A pivotal question in pandemic preparedness and therapeutic intervention is what are the specific mutations which are more likely to emerge during such global health crises? This study aims to identify markers for mutations with the highest probability of emergence in these pandemics, focusing on the SARS-CoV-2 spike protein, an essential and therapeutically significant viral protein, starting from sequence information from the onset of the pandemic until July 2022. Quite consistently, we observed that emerged mutations tended to demonstrate a high genetic score, which reflects high similarity of the type of codon required for translation between an amino acid and to the mutated one. Further, this pattern is also observed in therapeutically significant proteins of other ssRNA pandemic viruses, including influenza (HA, NA), spike proteins of Ebola, envelope of Dengue and Chikungunya. We propose that the genetic score serves as an initial indicator, preceding the actual impact of the mutation on viral fitness. Finally, we developed a comprehensive computational pipeline to further explore and predict the subsequent effects of mutations on viral fitness. We believe that our pipeline can narrow down and predict future mutations in therapeutically important viral proteins during a pandemic.
Collapse
Affiliation(s)
- Abhishek Sharma
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - C R Chandrashekar
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Sudhir Krishna
- National Centre for Biological Science, GKVK Campus, Bengaluru 560065, India
| | - Ramanathan Sowdhamini
- Molecular Biophysics Unit, Indian Institute of Science, Banagalore 560012, India; Institute of Bioinformatics and Applied Biotechnology, Bangalore, 560100, India.
| |
Collapse
|
4
|
da Silva Morgado F, Cahú R, de Jesus DC, de Souza Chaves LC, Ribeiro BM. Insect cell production of chimeric virus-like particles based on human immunodeficiency virus GAG proteins and yellow fever virus envelope protein. Braz J Microbiol 2024:10.1007/s42770-024-01509-5. [PMID: 39254800 DOI: 10.1007/s42770-024-01509-5] [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: 06/12/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The yellow fever virus (YFV) is a single stranded RNA virus belonging to the genus Orthoflavivirus that is capable of zoonotic transmissions that infect nonhuman and human primates. It is endemic in Brazil with recurrent epidemics of the disease, and it is transmitted through mosquitoes. The detection and immunization against YFV and other flaviviruses are fundamental for the management of the impacts of the disease in human environments. In an ongoing effort to develop new approaches for diagnostics and immunizations, we expressed VLPs displaying the yellow fever virus envelope protein (YFE) using recombinant baculovirus in insect cells. By co-expressing HIV-1 Pr55Gag protein (GAG) together with YFE we were able to generate chimeric VLPs containing a GAG core together with an envelope containing the YFE protein. The YFE and the chimeric GAG-YFE VLPs have potential as vaccine candidates and as reagents for serological assays in the detection of these viruses in human sera.
Collapse
Affiliation(s)
| | - Roberta Cahú
- Department of Cell Biology, Institute of Biology, University of Brasilia, Brasília, Brazil
| | | | | | | |
Collapse
|
5
|
Sukupayo PR, Poudel RC, Ghimire TR. Nature's Solution to Aedes Vectors: Toxorhynchites as a Biocontrol Agent. J Trop Med 2024; 2024:3529261. [PMID: 38948015 PMCID: PMC11213640 DOI: 10.1155/2024/3529261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 07/02/2024] Open
Abstract
This review summarizes the predatory potential of Toxorhynchites mosquitoes as biological control agents for Aedes vectors. A single larva can consume hundreds of mosquito larvae during its development, with preference for larger prey and higher consumption rates at higher prey densities. Studies suggest Toxorhynchites can significantly reduce Aedes populations. Beyond direct predation, they may indirectly influence prey behavior and adult mosquito lifespan. Despite the demonstrably positive effects of Toxorhynchites species as biocontrol agents, there are acknowledged limitations that require further investigation. These limitations include potential variations in effectiveness across diverse habitats and mosquito developmental stages. Additionally, long-term ecological sustainability and potential ramifications warrant further research. Future efforts should prioritize optimizing rearing and release strategies to enhance effectiveness. Exploring the potential for combined control methods with other biocontrol agents or traditional methods is also crucial. Finally, investigating the influence of environmental factors on predation rates can further refine and optimize the application of Toxorhynchites in mosquito control programs.
Collapse
Affiliation(s)
- Punya Ram Sukupayo
- Department of ZoologyBhaktapur Multiple CampusTribhuvan University, Bhaktapur, Nepal
- Central Department of ZoologyTribhuvan University, Kathmandu, Nepal
| | - Ram Chandra Poudel
- Molecular Biotechnology UnitNepal Academy of Science and Technology (NAST), Lalitpur, Nepal
| | - Tirth Raj Ghimire
- Department of ZoologyTri-Chandra Multiple CampusTribhuvan University, Kathmandu, Nepal
| |
Collapse
|
6
|
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. 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
|
7
|
Saivish MV, Nogueira ML, Rossi SL, Vasilakis N. Beyond Borders: Investigating the Mysteries of Cacipacoré, a Lesser-Studied Arbovirus in Brazil. Viruses 2024; 16:336. [PMID: 38543701 PMCID: PMC10975354 DOI: 10.3390/v16030336] [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: 02/02/2024] [Revised: 02/20/2024] [Accepted: 02/20/2024] [Indexed: 04/01/2024] Open
Abstract
Cacipacoré virus (CPCV) was discovered in 1977 deep in the Amazon rainforest from the blood of a black-faced ant thrush (Formicarius analis). As a member of the family Flaviviridae and genus orthoflavivirus, CPCV's intricate ecological association with vectors and hosts raises profound questions. CPCV's transmission cycle may involve birds, rodents, equids, bovines, marsupials, non-human primates, and bats as potential vertebrate hosts, whereas Culex and Aedes spp. mosquitoes have been implicated as potential vectors of transmission. The virus' isolation across diverse biomes, including urban settings, suggests its adaptability, as well as presents challenges for its accurate diagnosis, and thus its impact on veterinary and human health. With no specific treatment or vaccine, its prevention hinges on traditional arbovirus control measures. Here, we provide an overview of its ecology, transmission cycles, epidemiology, pathogenesis, and prevention, aiming at improving our ability to better understand this neglected arbovirus.
Collapse
Affiliation(s)
- Marielena V. Saivish
- Laboratórios de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, Sao Jose do Rio Preto 15090-000, SP, Brazil; (M.V.S.); (M.L.N.)
- Brazilian Biosciences National Laboratory, Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas 13083-100, SP, Brazil
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Maurício L. Nogueira
- Laboratórios de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto, Sao Jose do Rio Preto 15090-000, SP, Brazil; (M.V.S.); (M.L.N.)
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Shannan L. Rossi
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
| | - Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA;
- Center for Vector-Borne and Zoonotic Diseases, 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
| |
Collapse
|
8
|
Gabiane G, Bohers C, Mousson L, Obadia T, Dinglasan RR, Vazeille M, Dauga C, Viglietta M, Yébakima A, Vega-Rúa A, Gutiérrez Bugallo G, Gélvez Ramírez RM, Sonor F, Etienne M, Duclovel-Pame N, Blateau A, Smith-Ravin J, De Lamballerie X, Failloux AB. Evaluating vector competence for Yellow fever in the Caribbean. Nat Commun 2024; 15:1236. [PMID: 38336944 PMCID: PMC10858021 DOI: 10.1038/s41467-024-45116-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 01/16/2024] [Indexed: 02/12/2024] Open
Abstract
The mosquito-borne disease, Yellow fever (YF), has been largely controlled via mass delivery of an effective vaccine and mosquito control interventions. However, there are warning signs that YF is re-emerging in both Sub-Saharan Africa and South America. Imported from Africa in slave ships, YF was responsible for devastating outbreaks in the Caribbean. In Martinique, the last YF outbreak was reported in 1908 and the mosquito Aedes aegypti was incriminated as the main vector. We evaluated the vector competence of fifteen Ae. aegypti populations for five YFV genotypes (Bolivia, Ghana, Nigeria, Sudan, and Uganda). Here we show that mosquito populations from the Caribbean and the Americas were able to transmit the five YFV genotypes, with YFV strains for Uganda and Bolivia having higher transmission success. We also observed that Ae. aegypti populations from Martinique were more susceptible to YFV infection than other populations from neighboring Caribbean islands, as well as North and South America. Our vector competence data suggest that the threat of re-emergence of YF in Martinique and the subsequent spread to Caribbean nations and beyond is plausible.
Collapse
Affiliation(s)
- Gaelle Gabiane
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
- Université des Antilles, Ecole Doctorale 589, Schœlcher, Martinique, Marseille, France
| | - Chloé Bohers
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Laurence Mousson
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Thomas Obadia
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Marseille, France
- Institut Pasteur, Université Paris Cité, G5 Infectious Disease Epidemiology and Analytics, Paris, France
| | - Rhoel R Dinglasan
- University of Florida, Department of Infectious Diseases & Immunology and Emerging Pathogens Institute, College of Veterinary Medicine, Gainesville, FL, USA
| | - Marie Vazeille
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Catherine Dauga
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | - Marine Viglietta
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France
| | | | - Anubis Vega-Rúa
- Institut Pasteur de Guadeloupe, Laboratory of Vector Control Research, Unit Transmission Reservoir and Pathogens Diversity, Les Abymes, Guadeloupe, Marseille, France
| | - Gladys Gutiérrez Bugallo
- Institut Pasteur de Guadeloupe, Laboratory of Vector Control Research, Unit Transmission Reservoir and Pathogens Diversity, Les Abymes, Guadeloupe, Marseille, France
- Department of Vector Control, Center for Research, Diagnostic, and Reference, Institute of Tropical Medicine Pedro Kouri, Havana, Cuba
| | - Rosa Margarita Gélvez Ramírez
- Centro de Atención y Diagnóstico de Enfermedades Infecciosas, Fundación INFOVIDA, Bucaramanga, Colombia
- Unité des Virus Emergents (UVE), Aix Marseille Université, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Fabrice Sonor
- Centre de Démoustication et de Recherches Entomologiques, Lutte antivectorielle, Martinique, Marseille, France
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Manuel Etienne
- Centre de Démoustication et de Recherches Entomologiques, Lutte antivectorielle, Martinique, Marseille, France
| | - Nathalie Duclovel-Pame
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Alain Blateau
- Agence Régionale de Santé, Direction de la Santé Publique, Martinique, Marseille, France
| | - Juliette Smith-Ravin
- Groupe de recherche Biospheres Université des Antilles, Campus de Schœlcher, Martinique, Marseille, France
| | - Xavier De Lamballerie
- Unité des Virus Emergents (UVE), Aix Marseille Université, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Anna-Bella Failloux
- Institut Pasteur, Université Paris Cité, Arboviruses and Insect Vectors, Paris, France.
| |
Collapse
|
9
|
Serafim MSM, Kronenberger T, Rocha REO, Rosa ADRA, Mello TLG, Poso A, Ferreira RS, Abrahão JS, Kroon EG, Mota BEF, Maltarollo VG. Aminopyrimidine Derivatives as Multiflavivirus Antiviral Compounds Identified from a Consensus Virtual Screening Approach. J Chem Inf Model 2024; 64:393-411. [PMID: 38194508 DOI: 10.1021/acs.jcim.3c01505] [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: 01/11/2024]
Abstract
Around three billion people are at risk of infection by the dengue virus (DENV) and potentially other flaviviruses. Worldwide outbreaks of DENV, Zika virus (ZIKV), and yellow fever virus (YFV), the lack of antiviral drugs, and limitations on vaccine usage emphasize the need for novel antiviral research. Here, we propose a consensus virtual screening approach to discover potential protease inhibitors (NS3pro) against different flavivirus. We employed an in silico combination of a hologram quantitative structure-activity relationship (HQSAR) model and molecular docking on characterized binding sites followed by molecular dynamics (MD) simulations, which filtered a data set of 7.6 million compounds to 2,775 hits. Lastly, docking and MD simulations selected six final potential NS3pro inhibitors with stable interactions along the simulations. Five compounds had their antiviral activity confirmed against ZIKV, YFV, DENV-2, and DENV-3 (ranging from 4.21 ± 0.14 to 37.51 ± 0.8 μM), displaying aggregator characteristics for enzymatic inhibition against ZIKV NS3pro (ranging from 28 ± 7 to 70 ± 7 μM). Taken together, the compounds identified in this approach may contribute to the design of promising candidates to treat different flavivirus infections.
Collapse
Affiliation(s)
- Mateus Sá Magalhães Serafim
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), Tübingen 72076, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
| | - Rafael Eduardo Oliveira Rocha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Amanda Del Rio Abreu Rosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Thaysa Lara Gonçalves Mello
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Antti Poso
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, Tübingen 72076, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Department of Medical Oncology and Pneumology, University Hospital of Tübingen, Tübingen 70211, Germany
| | - Rafaela Salgado Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Jonatas Santos Abrahão
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Erna Geessien Kroon
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Bruno Eduardo Fernandes Mota
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 31270-901, Brazil
| |
Collapse
|
10
|
Sotorilli GE, Gravina HD, de Carvalho AC, Shimizu JF, Fontoura MA, Melo-Hanchuk TD, Cordeiro AT, Marques RE. Phenotypical Screening of an MMV Open Box Library and Identification of Compounds with Antiviral Activity against St. Louis Encephalitis Virus. Viruses 2023; 15:2416. [PMID: 38140657 PMCID: PMC10747599 DOI: 10.3390/v15122416] [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: 10/14/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
St. Louis encephalitis virus (SLEV) is a neglected mosquito-borne Flavivirus that may cause severe neurological disease in humans and other animals. There are no specific treatments against SLEV infection or disease approved for human use, and drug repurposing may represent an opportunity to accelerate the development of treatments against SLEV. Here we present a scalable, medium-throughput phenotypic cell culture-based screening assay on Vero CCL81 cells to identify bioactive compounds that could be repurposed against SLEV infection. We screened eighty compounds from the Medicines for Malaria Venture (MMV) COVID Box library to identify nine (11%) compounds that protected cell cultures from SLEV-induced cytopathic effects, with low- to mid-micromolar potencies. We validated six hit compounds using viral plaque-forming assays to find that the compounds ABT-239, Amiodarone, Fluphenazine, Posaconazole, Triparanol, and Vidofludimus presented varied levels of antiviral activity and selectivity depending on the mammalian cell type used for testing. Importantly, we identified and validated the antiviral activity of the anti-flavivirus nucleoside analog 7DMA against SLEV. Triparanol and Fluphenazine reduced infectious viral loads in both Vero CCL81 and HBEC-5i cell cultures and, similar to the other validated compounds, are likely to exert antiviral activity through a molecular target in the host.
Collapse
Affiliation(s)
- Giuliana Eboli Sotorilli
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Genetics, Microbiology and Immunology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Humberto Doriguetto Gravina
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Ana Carolina de Carvalho
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Genetics, Microbiology and Immunology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Jacqueline Farinha Shimizu
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Marina Alves Fontoura
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
- Department of Cellular and Structural Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas 13083-100, Brazil
| | - Talita Diniz Melo-Hanchuk
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Artur Torres Cordeiro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, Brazil; (G.E.S.); (H.D.G.); (A.C.d.C.); (J.F.S.); (M.A.F.); (T.D.M.-H.); (A.T.C.)
| |
Collapse
|
11
|
Affiliation(s)
- Peter J Hotez
- From the Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston (P.J.H.); and the Department of Pediatrics, Stanford University School of Medicine, Stanford, CA (A.D.L.)
| | - Angelle Desiree LaBeaud
- From the Departments of Pediatrics and Molecular Virology and Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston (P.J.H.); and the Department of Pediatrics, Stanford University School of Medicine, Stanford, CA (A.D.L.)
| |
Collapse
|
12
|
Chen GH, Dai YC, Hsieh SC, Tsai JJ, Sy AK, Jiz M, Pedroso C, Brites C, Netto EM, Kanki PJ, Saunders DRD, Vanlandingham DL, Higgs S, Huang YJS, Wang WK. Detection of anti-premembrane antibody as a specific marker of four flavivirus serocomplexes and its application to serosurveillance in endemic regions. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.21.23295701. [PMID: 37808865 PMCID: PMC10557774 DOI: 10.1101/2023.09.21.23295701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
In the past few decades, several emerging/re-emerging mosquito-borne flaviviruses have resulted in disease outbreaks of public health concern in the tropics and subtropics. Due to cross-reactivities of antibodies recognizing the envelope protein of different flaviviruses, serosurveillance remains a challenge. Previously we reported that anti-premembrane (prM) antibody can discriminate between three flavivirus infections by Western blot analysis. In this study, we aimed to develop a serological assay that can discriminate infection or exposure with flaviviruses from four serocomplexes, including dengue (DENV), Zika (ZIKV), West Nile (WNV) and yellow fever (YFV) viruses, and explore its application for serosurveillance in flavivirus-endemic countries. We employed Western blot analysis including antigens of six flaviviruses (DENV1, 2 and 4, WNV, ZIKV and YFV) from four serocomplexes. We tested serum samples from YF-17D vaccinees, and from DENV, ZIKV and WNV panels that had been confirmed by RT-PCR or by neutralization assays. The overall sensitivity/specificity of anti-prM antibodies for DENV, ZIKV, WNV, and YFV infections/exposure were 91.7%/96.4%, 91.7%/99.2%, 88.9%/98.3%, and 91.3%/92.5%, respectively. When testing 48 samples from Brazil, we identified multiple flavivirus infections/exposure including DENV and ZIKV, DENV and YFV, and DENV, ZIKV and YFV. When testing 50 samples from the Philippines, we detected DENV, ZIKV, and DENV and ZIKV infections with a ZIKV seroprevalence rate of 10%, which was consistent with reports of low-level circulation of ZIKV in Asia. Together, these findings suggest that anti-prM antibody is a flavivirus serocomplex-specific marker and can be employed to delineate four flavivirus infections/exposure in regions where multiple flaviviruses co-circulate.
Collapse
|
13
|
Lippi CA, Mundis SJ, Sippy R, Flenniken JM, Chaudhary A, Hecht G, Carlson CJ, Ryan SJ. Trends in mosquito species distribution modeling: insights for vector surveillance and disease control. Parasit Vectors 2023; 16:302. [PMID: 37641089 PMCID: PMC10463544 DOI: 10.1186/s13071-023-05912-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023] Open
Abstract
Species distribution modeling (SDM) has become an increasingly common approach to explore questions about ecology, geography, outbreak risk, and global change as they relate to infectious disease vectors. Here, we conducted a systematic review of the scientific literature, screening 563 abstracts and identifying 204 studies that used SDMs to produce distribution estimates for mosquito species. While the number of studies employing SDM methods has increased markedly over the past decade, the overwhelming majority used a single method (maximum entropy modeling; MaxEnt) and focused on human infectious disease vectors or their close relatives. The majority of regional models were developed for areas in Africa and Asia, while more localized modeling efforts were most common for North America and Europe. Findings from this study highlight gaps in taxonomic, geographic, and methodological foci of current SDM literature for mosquitoes that can guide future efforts to study the geography of mosquito-borne disease risk.
Collapse
Affiliation(s)
- Catherine A Lippi
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, 32601, USA.
| | - Stephanie J Mundis
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA
| | - Rachel Sippy
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA
- School of Mathematics and Statistics, University of St Andrews, St Andrews, KY16 9SS, UK
| | - J Matthew Flenniken
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA
| | - Anusha Chaudhary
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA
| | - Gavriella Hecht
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, 32601, USA
| | - Colin J Carlson
- Center for Global Health Science and Security, Georgetown University Medical Center, Georgetown University, Washington, DC, USA
| | - Sadie J Ryan
- Quantitative Disease Ecology and Conservation (QDEC) Lab, Department of Geography, University of Florida, Gainesville, FL, 32601, USA.
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, 32601, USA.
| |
Collapse
|
14
|
Tubaki RM, de Menezes RMT, David MR, Palasio RGS, de Aguiar OT, Baitello JB, Santos VO, Balbino N, Chiaravalloti-Neto F. Physical Attributes of Tree Holes in the Atlantic Forest Edges: Evaluating Their Association with the Presence and Abundance of Immature Haemagogus leucocelaenus. Trop Med Infect Dis 2023; 8:337. [PMID: 37505633 PMCID: PMC10383151 DOI: 10.3390/tropicalmed8070337] [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: 05/04/2023] [Revised: 06/09/2023] [Accepted: 06/18/2023] [Indexed: 07/29/2023] Open
Abstract
Sylvatic yellow fever (SYF) was recently a health issue in Brazil (2016-2019) because transmission was facilitated by a high density of vectors, amplifying hosts, and low vaccine coverage of the human population, especially in urban forests in the Southeast Region of Brazil. Moreover, urban forest edges are more likely to have contact between human and sylvatic vector mosquito populations. Here, we show the association between abiotic and biotic features of tree holes as Haemagogus leucocelaenus rearing sites in Cantareira State Park in Atlantic Forest edges. The analyzed physical features of the tree holes were diameter at breast height, tree hole opening diameter, depth, trunk diameter, tree hole volume, collected volume, height (varying from 0.02 to 4.2 m above ground), and the presence of Culicidae species other than Hg. leucocelaenus. We analyzed 105 positive and 68 negative water samples for larval presence and found no differences between them, suggesting the lack of specific physical characteristics in these categories. Hg. leucocelaenus larval abundance was correlated with the collected volume and opening diameter of tree holes. The tree species that most represented negative breeding sites were Euplassa cantareirae, Guarea macrophylla, Psychotria suterella, and Tibouchina pulchra. Four significant clusters as areas with a high risk of SYV were identified by Get-Ordis spatial analysis. Although Hg. leucocelaenus larvae were found in tree holes with high water levels, their occurrence was regulated by that of other mosquito species. Our findings contribute to clarifying immature vector ecology in tree holes related to human exposure to SYF in urban forest edges.
Collapse
Affiliation(s)
- Rosa Maria Tubaki
- Laboratório de Entomologia Médica, Instituto Pasteur da Secretaria Estadual de Saúde de São Paulo, São Paulo 01027-000, Brazil
| | - Regiane Maria Tironi de Menezes
- Laboratório de Entomologia Médica, Instituto Pasteur da Secretaria Estadual de Saúde de São Paulo, São Paulo 01027-000, Brazil
| | - Mariana Rocha David
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Rio de Janeiro 17700-000, Brazil
| | | | - Osny Tadeu de Aguiar
- Instituto de Pesquisas Ambientais, Secretaria de Infraestrutura e Meio Ambiente, São Paulo 90690-000, Brazil
| | - João Batista Baitello
- Instituto de Pesquisas Ambientais, Secretaria de Infraestrutura e Meio Ambiente, São Paulo 90690-000, Brazil
| | - Vagner Oliveira Santos
- Superintendência de Controle de Endemias, Secretária Estadual da Saúde, São Paulo 74605-110, Brazil
| | - Natália Balbino
- Superintendência de Controle de Endemias, Secretária Estadual da Saúde, São Paulo 74605-110, Brazil
| | - Francisco Chiaravalloti-Neto
- Departamento de Epidemiologia, Faculdade de Saúde Pública, Universidade de São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
15
|
Pawlak JB, Hsu JCC, Xia H, Han P, Suh HW, Grove TL, Morrison J, Shi PY, Cresswell P, Laurent-Rolle M. CMPK2 restricts Zika virus replication by inhibiting viral translation. PLoS Pathog 2023; 19:e1011286. [PMID: 37075076 PMCID: PMC10150978 DOI: 10.1371/journal.ppat.1011286] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 05/01/2023] [Accepted: 03/09/2023] [Indexed: 04/20/2023] Open
Abstract
Flaviviruses continue to emerge as global health threats. There are currently no Food and Drug Administration (FDA) approved antiviral treatments for flaviviral infections. Therefore, there is a pressing need to identify host and viral factors that can be targeted for effective therapeutic intervention. Type I interferon (IFN-I) production in response to microbial products is one of the host's first line of defense against invading pathogens. Cytidine/uridine monophosphate kinase 2 (CMPK2) is a type I interferon-stimulated gene (ISG) that exerts antiviral effects. However, the molecular mechanism by which CMPK2 inhibits viral replication is unclear. Here, we report that CMPK2 expression restricts Zika virus (ZIKV) replication by specifically inhibiting viral translation and that IFN-I- induced CMPK2 contributes significantly to the overall antiviral response against ZIKV. We demonstrate that expression of CMPK2 results in a significant decrease in the replication of other pathogenic flaviviruses including dengue virus (DENV-2), Kunjin virus (KUNV) and yellow fever virus (YFV). Importantly, we determine that the N-terminal domain (NTD) of CMPK2, which lacks kinase activity, is sufficient to restrict viral translation. Thus, its kinase function is not required for CMPK2's antiviral activity. Furthermore, we identify seven conserved cysteine residues within the NTD as critical for CMPK2 antiviral activity. Thus, these residues may form an unknown functional site in the NTD of CMPK2 contributing to its antiviral function. Finally, we show that mitochondrial localization of CMPK2 is required for its antiviral effects. Given its broad antiviral activity against flaviviruses, CMPK2 is a promising potential pan-flavivirus inhibitor.
Collapse
Affiliation(s)
- Joanna B. Pawlak
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jack Chun-Chieh Hsu
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hongjie Xia
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Patrick Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Hee-Won Suh
- Department of Biomedical Engineering, Yale University School of Engineering and Applied Science, New Haven, Connecticut, United States of America
| | - Tyler L. Grove
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Juliet Morrison
- Department of Microbiology and Plant Pathology, University of California, Riverside, California, United States of America
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Maudry Laurent-Rolle
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
16
|
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: 0] [Impact Index Per Article: 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
|
17
|
Immunogenicity and protective activity of mRNA vaccine candidates against yellow fever virus in animal models. NPJ Vaccines 2023; 8:31. [PMID: 36871059 PMCID: PMC9984760 DOI: 10.1038/s41541-023-00629-7] [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: 07/15/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Despite the success of the widely used attenuated yellow fever (YF) vaccine, its global supply remains a substantial barrier to implementing vaccination campaigns in endemic regions and combating emerging epidemics. In A129 mice and rhesus macaques, we evaluated the immunogenicity and protective activity of messenger RNA (mRNA) vaccine candidates encapsulated in lipid nanoparticles, expressing the pre-membrane and envelope proteins or the non-structural protein 1 of YF virus. Vaccine constructs induced humoral and cell-mediated immune responses in mice, resulting in protection against lethal YF virus infection after passive administration of serum or splenocytes from vaccinated mice. Vaccination of macaques induced sustained high humoral and cellular immune responses for at least 5 months after the second dose. Our data demonstrate that these mRNA vaccine candidates can be considered an attractive addition to the licensed YF vaccine supply based on the induction of functional antibodies correlating with protection and T-cell responses; they could alleviate the limited supply of current YF vaccines, mitigating future YF epidemics.
Collapse
|
18
|
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: 2] [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
|
19
|
Ribeiro IP, Delatorre E, de Abreu FVS, dos Santos AAC, Furtado ND, Ferreira-de-Brito A, de Pina-Costa A, Neves MSAS, de Castro MG, Motta MDA, Brasil P, Lourenço-de-Oliveira R, Bonaldo MC. Ecological, Genetic, and Phylogenetic Aspects of YFV 2017-2019 Spread in Rio de Janeiro State. Viruses 2023; 15:v15020437. [PMID: 36851651 PMCID: PMC9961572 DOI: 10.3390/v15020437] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
In Brazil, a yellow fever (YF) outbreak was reported in areas considered YF-free for decades. The low vaccination coverage and the increasing forest fragmentation, with the wide distribution of vector mosquitoes, have been related to yellow fever virus (YFV) transmission beyond endemic areas since 2016. Aiming to elucidate the molecular and phylogenetic aspects of YFV spread on a local scale, we generated 43 new YFV genomes sampled from humans, non-human primates (NHP), and primarily, mosquitoes from highly heterogenic areas in 15 localities from Rio de Janeiro (RJ) state during the YFV 2016-2019 outbreak in southeast Brazil. Our analysis revealed that the genetic diversity and spatial distribution of the sylvatic transmission of YFV in RJ originated from at least two introductions and followed two chains of dissemination, here named the YFV RJ-I and YFV RJ-II clades. They moved with similar dispersal speeds from the north to the south of the RJ state in parallel directions, separated by the Serra do Mar Mountain chain, with YFV RJ-I invading the north coast of São Paulo state. The YFV RJ-I clade showed a more significant heterogeneity across the entire polyprotein. The YFV RJ-II clade, with only two amino acid polymorphisms, mapped at NS1 (I1086V), present only in mosquitoes at the same locality and NS4A (I2176V), shared by all YFV clade RJ-II, suggests a recent clustering of YFV isolates collected from different hosts. Our analyses strengthen the role of surveillance, genomic analyses of YVF isolated from other hosts, and environmental studies into the strategies to forecast, control, and prevent yellow fever outbreaks.
Collapse
Affiliation(s)
- Ieda Pereira Ribeiro
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Edson Delatorre
- Laboratório de Genômica Evolutiva e Ambiental, Departamento de Biologia, Centro de Ciências Exatas, Naturais e da Saúde, Universidade Federal do Espírito Santo, Alegre 29500-000, ES, Brazil
| | - Filipe Vieira Santos de Abreu
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
- Instituto Federal do Norte de Minas Gerais, Salinas 39560-000, MG, Brazil
| | - Alexandre Araújo Cunha dos Santos
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Nathália Dias Furtado
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Anielly Ferreira-de-Brito
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Anielle de Pina-Costa
- Laboratório de Doenças Febris Agudas, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
- Faculdade de Medicina de Teresópolis, Centro Universitário Serra dos Órgãos, UNIFESO, Teresópolis 25955-001, RJ, Brazil
| | | | - Márcia Gonçalves de Castro
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Monique de Albuquerque Motta
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Patricia Brasil
- Laboratório de Doenças Febris Agudas, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
| | - Ricardo Lourenço-de-Oliveira
- Laboratório de Mosquitos Transmissores de Hematozoários, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
- Correspondence: (R.L.-d.-O.); (M.C.B.)
| | - Myrna Cristina Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21040-900, RJ, Brazil
- Correspondence: (R.L.-d.-O.); (M.C.B.)
| |
Collapse
|
20
|
Ferreira LS, Darcie Marquitti FM, Paixão da Silva RL, Borges ME, Ferreira da Costa Gomes M, Cruz OG, Kraenkel RA, Coutinho RM, Prado PI, Bastos LS. Estimating the impact of implementation and timing of the COVID-19 vaccination programme in Brazil: a counterfactual analysis. LANCET REGIONAL HEALTH. AMERICAS 2023; 17:100397. [PMID: 36439909 PMCID: PMC9676113 DOI: 10.1016/j.lana.2022.100397] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/30/2022] [Accepted: 10/27/2022] [Indexed: 11/22/2022]
Abstract
Background Vaccines developed between 2020 and 2021 against the SARS-CoV-2 virus were designed to diminish the severity and prevent deaths due to COVID-19. However, estimates of the effectiveness of vaccination campaigns in achieving these goals remain a methodological challenge. In this work, we developed a Bayesian statistical model to estimate the number of deaths and hospitalisations averted by vaccination of older adults (above 60 years old) in Brazil. Methods We fit a linear model to predict the number of deaths and hospitalisations of older adults as a function of vaccination coverage in this group and casualties in younger adults. We used this model in a counterfactual analysis, simulating alternative scenarios without vaccination or with faster vaccination roll-out. We estimated the direct effects of COVID-19 vaccination by computing the difference between hypothetical and realised scenarios. Findings We estimated that more than 165,000 individuals above 60 years of age were not hospitalised due to COVID-19 in the first seven months of the vaccination campaign. An additional contingent of 104,000 hospitalisations could have been averted if vaccination had started earlier. We also estimated that more than 58 thousand lives were saved by vaccinations in the period analysed for the same age group and that an additional 47 thousand lives could have been saved had the Brazilian government started the vaccination programme earlier. Interpretation Our estimates provided a lower bound for vaccination impacts in Brazil, demonstrating the importance of preventing the suffering and loss of older Brazilian adults. Once vaccines were approved, an early vaccination roll-out could have saved many more lives, especially when facing a pandemic. Funding The Coordenação de Aperfeiçoamento de Pessoal de Nível Superior-Brazil (Finance Code 001 to F.M.D.M. and L.S.F.), Conselho Nacional de Desenvolvimento Científico e Tecnológico - Brazil (grant number: 315854/2020-0 to M.E.B., 141698/2018-7 to R.L.P.d.S., 313055/2020-3 to P.I.P., 311832/2017-2 to R.A.K.), Fundação de Amparo à Pesquisa do Estado de São Paulo - Brazil (contract number: 2016/01343-7 to R.A.K.), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro - Brazil (grant number: E-26/201.277/2021 to L.S.B.) and Inova Fiocruz/Fundação Oswaldo Cruz - Brazil (grant number: 48401485034116) to L.S.B., O.G.C. and M.G.d.F.C. The funding agencies had no role in the conceptualization of the study.
Collapse
Affiliation(s)
- Leonardo Souto Ferreira
- Instituto de Física Teórica, Universidade Estadual Paulista, São Paulo, Brazil
- Observatório COVID-19 BR, Brazil
| | - Flavia Maria Darcie Marquitti
- Observatório COVID-19 BR, Brazil
- Instituto de Física ‘Gleb Wataghin’ and Instituto de Biologia, Universidade Estadual de Campinas, Campinas, Brazil
| | | | | | | | - Oswaldo Gonçalves Cruz
- Observatório COVID-19 BR, Brazil
- Fundação Oswaldo Cruz, Programa de Computação Científica, Rio de Janeiro, Brazil
| | - Roberto André Kraenkel
- Instituto de Física Teórica, Universidade Estadual Paulista, São Paulo, Brazil
- Observatório COVID-19 BR, Brazil
| | - Renato Mendes Coutinho
- Observatório COVID-19 BR, Brazil
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, Santo André, Brazil
| | - Paulo Inácio Prado
- Observatório COVID-19 BR, Brazil
- Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Leonardo Soares Bastos
- Observatório COVID-19 BR, Brazil
- Fundação Oswaldo Cruz, Programa de Computação Científica, Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Cajaraville ACDRA, Gomes MPDB, Azamor T, Pereira RC, Neves PCDC, De Luca PM, de Lima SMB, Gaspar LP, Caride E, Freire MDS, Medeiros MA. Evaluation of Two Adjuvant Formulations for an Inactivated Yellow Fever 17DD Vaccine Candidate in Mice. Vaccines (Basel) 2022; 11:73. [PMID: 36679918 PMCID: PMC9865672 DOI: 10.3390/vaccines11010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/10/2022] [Accepted: 12/24/2022] [Indexed: 12/31/2022] Open
Abstract
The attenuated yellow fever (YF) vaccine is one of the most successful vaccines ever developed. After a single dose administration YF vaccine can induce balanced Th1/Th2 immune responses and long-lasting neutralizing antibodies. These attributes endorsed it as a model of how to properly stimulate the innate response to target protective immune responses. Despite their longstanding success, attenuated YF vaccines can cause rare fatal adverse events and are contraindicated for persons with immunosuppression, egg allergy and age < 6 months and >60 years. These drawbacks have encouraged the development of a non-live vaccine. The aim of the present study is to characterize and compare the immunological profile of two adjuvant formulations of an inactivated YF 17DD vaccine candidate. Inactivated YF vaccine formulations based on alum (Al(OH)3) or squalene (AddaVax®) were investigated by immunization of C57BL/6 mice in 3-dose or 2-dose schedules, respectively, and compared with a single dose of attenuated YF virus 17DD. Sera were analyzed by ELISA and Plaque Reduction Neutralization Test (PRNT) for detection of total IgG and neutralizing antibodies against YF virus. In addition, splenocytes were collected to evaluate cellular responses by ELISpot. Both inactivated formulations were able to induce high titers of IgG against YF, although neutralizing antibodies levels were borderline on pre-challenge samples. Analysis of IgG subtypes revealed a predominance of IgG2a associated with improved neutralizing capacity in animals immunized with the attenuated YF vaccine, and a predominance of IgG1 in groups immunized with experimental non-live formulations (alum and AddaVax®). After intracerebral (IC) challenge, attenuated and inactivated vaccine formulations showed an increase in neutralizing antibodies. The AddaVax®-based inactivated vaccine and the attenuated vaccine achieved 100% protection, and alum-based equivalent formulation achieved 70% protection.
Collapse
Affiliation(s)
| | - Mariana Pierre de Barros Gomes
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Tamiris Azamor
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Renata Carvalho Pereira
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Patrícia Cristina da Costa Neves
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Paula Mello De Luca
- Instituto Oswaldo Cruz (IOC), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Sheila Maria Barbosa de Lima
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Luciane Pinto Gaspar
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Elena Caride
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marcos da Silva Freire
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| | - Marco Alberto Medeiros
- Vice Diretoria de Desenvolvimento Tecnológico (VDTEC), Instituto de Tecnologia em Imunobiológicos (Bio-Manguinhos), FIOCRUZ Av. Brasil, Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
22
|
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: 1.5] [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
|
23
|
Development of a Bicistronic Yellow Fever Live Attenuated Vaccine with Reduced Neurovirulence and Viscerotropism. Microbiol Spectr 2022; 10:e0224622. [PMID: 35980184 PMCID: PMC9602263 DOI: 10.1128/spectrum.02246-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The yellow fever (YF) live attenuated vaccine strain 17D (termed 17D) has been widely used for the prevention and control of YF disease. However, 17D retains significant neurovirulence and viscerotropism in mice, which is probably linked to the increased occurrences of serious adverse events following 17D vaccination. Thus, the development of an updated version of the YF vaccine with an improved safety profile is of high priority. Here, we generated a viable bicistronic YF virus (YFV) by incorporating the internal ribosome entry site (IRES) from Encephalomyocarditis virus into an infectious clone of YFV 17D. The resulting recombinant virus, 17D-IRES, exhibited similar replication efficiency to its parental virus (17D) in mammalian cell lines, while it was highly restricted in mosquito cells. Serial passage of 17D-IRES in BHK-21 cells showed good genetic stability. More importantly, in comparison with the parental 17D, 17D-IRES displayed significantly decreased mouse neurovirulence and viscerotropism in type I interferon (IFN)-signaling-deficient and immunocompetent mouse models. Interestingly, 17D-IRES showed enhanced sensitivity to type I IFN compared with 17D. Moreover, immunization with 17D-IRES provided solid protection for mice against a lethal challenge with YFV. These preclinical data support further development of 17D-IRES as an updated version for the approved YF vaccine. This IRES-based attenuation strategy could be also applied to the design of live attenuated vaccines against other mosquito-borne flaviviruses. IMPORTANCE Yellow fever (YF) continually spreads and causes epidemics around the world, posing a great threat to human health. The YF live attenuated vaccine 17D is considered the most efficient vaccine available and helps to successfully control disease epidemics. However, side effects may occur after vaccination, such as viscerotropic disease (YEL-AVD) and neurotropic adverse disease (YEL-AND). Thus, there is an urgent need for a safer YF vaccine. Here, an IRES strategy was employed, and a bicistronic YFV was successfully developed (named 17D-IRES). 17D-IRES showed effective replication and genetic stability in vitro and high attenuation in vivo. Importantly, 17D-IRES induced humoral and cellular immune responses and conferred full protection against lethal YFV challenge. Our study provides data suggesting that 17D-IRES, with its prominent advantages, could be a vaccine candidate against YF. Moreover, this IRES-based bicistronic technology platform represents a promising strategy for developing other live attenuated vaccines against emerging viruses.
Collapse
|
24
|
Basile AJ, Niedrig M, Lambert AJ, Meurant R, Brault AC, Domingo C, Goodman CH, Johnson BW, Mossel EC, Mulders MN, Velez JO, Hughes HR. Laboratory evaluation of RealStar Yellow Fever Virus RT-PCR kit 1.0 for potential use in the global yellow fever laboratory network. PLoS Negl Trop Dis 2022; 16:e0010770. [PMID: 36067233 PMCID: PMC9481164 DOI: 10.1371/journal.pntd.0010770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/16/2022] [Accepted: 08/26/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Early detection of human yellow fever (YF) infection in YF-endemic regions is critical to timely outbreak mitigation. African National Laboratories chiefly rely on serological assays that require confirmation at Regional Reference Laboratories, thus delaying results, which themselves are not always definitive often due to antibody cross-reactivity. A positive molecular test result is confirmatory for YF; therefore, a standardized YF molecular assay would facilitate immediate confirmation at National Laboratories. The WHO-coordinated global Eliminate Yellow Fever Epidemics Laboratory Technical Working Group sought to independently evaluate the quality and performance of commercial YF molecular assays relevant to use in countries with endemic YF, in the absence of stringent premarket assessments. This report details a limited laboratory WHO-coordinated evaluation of the altona Diagnostics RealStar Yellow Fever Virus RT-PCR kit 1.0.
Methodology and principal findings
Specific objectives were to assess the assay’s ability to detect YF virus strains in human serum from YF-endemic regions, determine the potential for interference and cross-reactions, verify the performance claims as stated by the manufacturer, and assess usability. RNA extracted from normal human serum spiked with YF virus showed the assay to be precise with minimal lot-to-lot variation. The 95% limit of detection calculated was approximately 1,245 RNA copies/ml [95% confidence interval 497 to 1,640 copies/ml]. Positive results were obtained with spatially and temporally diverse YF strains. The assay was specific for YF virus, was not subject to endogenous or exogenous interferents, and was clinically sensitive and specific. A review of operational characteristics revealed that a positivity cutoff was not defined in the instructions for use, but otherwise the assay was user-friendly.
Conclusions and significance
The RealStar Yellow Fever Virus RT-PCR kit 1.0 has performance characteristics consistent with the manufacturer’s claims and is suitable for use in YF-endemic regions. Its use is expected to decrease YF outbreak detection times and be instrumental in saving lives.
Collapse
Affiliation(s)
- Alison J. Basile
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
- * E-mail: (AJB); (HRH)
| | | | - Amy J. Lambert
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | | | - Aaron C. Brault
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Cristina Domingo
- Public Health Laboratory Support Unit, Centre for International Health Protection, Robert Koch Institute, Berlin, Germany
| | - Christin H. Goodman
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Barbara W. Johnson
- Scientific Laboratory Consulting, Laporte, Colorado, United States of America
| | - Eric C. Mossel
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | | | - Jason O. Velez
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
| | - Holly R. Hughes
- Arboviral Diseases Branch, Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, United States of America
- * E-mail: (AJB); (HRH)
| |
Collapse
|
25
|
Müller GA, de Mello CF, Bueno AS, de Alcantara Azevedo WT, Alencar J. Little noticed, but very important: The role of breeding sites formed by bamboos in maintaining the diversity of mosquitoes (Diptera: Culicidae) in the Atlantic Forest biome. PLoS One 2022; 17:e0273774. [PMID: 36067179 PMCID: PMC9447929 DOI: 10.1371/journal.pone.0273774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/12/2022] [Indexed: 11/18/2022] Open
Abstract
This study investigated the composition of mosquito species in different kinds of breeding sites in a tropical forest remnant of the Atlantic Forest and identified species of public health concern therein. Collections of immature forms of mosquitoes were carried out monthly at the Poço das Antas Biological Reserve in southeastern Brazil, between June 2014 and June 2015. Samples were collected from four types of breeding sites: bamboos, bromeliads, puddles, and a lake. A total of 1,182 specimens of mosquitoes belonging to 28 species and 13 genera were collected. Three species, Ad. squamipennis, An. neglectus, and Wy. arthrostigma represented 64.8% of the captured specimens. Only three species were found in more than one type of breeding site: Ps. ferox, An. triannulatus, and Tx. trichopygus. Two species of public health concern were found breeding in bamboo (Ae. aegypti and Ae. albopictus) and one in the lake (An. darlingi). Bamboo had the highest species richness, Shannon diversity, abundance of individuals and number of dominant species of all breeding sites. Similar Simpson diversity was obtained for bamboo and bromeliads, with higher values than those obtained for puddles and the lake. The significance of the four breeding sites, especially bamboos, is discussed in the context of controlling populations of sylvatic species of mosquitoes in Atlantic Forest areas.
Collapse
Affiliation(s)
- Gerson Azulim Müller
- Instituto Federal de Educação, Ciência e Tecnologia Farroupilha, Panambi, RS, Brazil
| | - Cecilia Ferreira de Mello
- Laboratório de Diptera, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação em Biologia Animal, Instituto de Biologia (UFRRJ), Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Anderson S. Bueno
- Instituto Federal de Educação, Ciência e Tecnologia Farroupilha, Júlio de Castilhos, RS, Brazil
| | - Wellington Thadeu de Alcantara Azevedo
- Laboratório de Diptera, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Programa de Pós-Graduação em Biologia Animal, Instituto de Biologia (UFRRJ), Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - Jeronimo Alencar
- Laboratório de Diptera, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- * E-mail:
| |
Collapse
|
26
|
Phenotypic and Genetic Studies of the Viral Lineage Associated with the Recent Yellow Fever Outbreak in Brazil. Viruses 2022; 14:v14081818. [PMID: 36016440 PMCID: PMC9412561 DOI: 10.3390/v14081818] [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: 07/08/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Yellow fever virus (YFV) caused an outbreak in the Brazilian Southeast from 2016 to 2019, of the most significant magnitude since the 1900s. An investigation of the circulating virus revealed that most of the genomes detected in this period carried nine unique amino acid polymorphisms, with eight located in the non-structural proteins NS3 and NS5, which are pivotal for viral replication. To elucidate the effect of these amino acid changes on viral infection, we constructed viruses carrying amino acid alterations in NS3 and NS5, performed infection in different cells, and assessed their neurovirulence in BALB/c mice and infected AG129 mice. We observed that the residues that compose the YFV 2016-2019 molecular signature in the NS5 protein might have been related to an attenuated phenotype, and that the alterations in the NS3 protein only slightly affected viral infection in AG129 mice, increasing to a low extent the mortality rate of these animals. These results contributed to unveiling the role of specific naturally occurring amino acid changes in the circulating strain of YFV in Brazil.
Collapse
|
27
|
Mapping and Validation of Peptides Differentially Recognized by Antibodies from the Serum of Yellow Fever Virus-Infected or 17DD-Vaccinated Patients. Viruses 2022; 14:v14081645. [PMID: 36016268 PMCID: PMC9415205 DOI: 10.3390/v14081645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/12/2022] [Accepted: 07/25/2022] [Indexed: 02/01/2023] Open
Abstract
Yellow Fever disease is caused by the Yellow Fever virus (YFV), an arbovirus from the Flaviviridae family. The re-emergence of Yellow Fever (YF) was facilitated by the increasing urbanization of sylvatic areas, the wide distribution of the mosquito vector, and the low percentage of people immunized in the Americas, which caused severe outbreaks in recent years, with a high mortality rate. Therefore, serological approaches capable of discerning antibodies generated from the wild-type (YFV-WT) strain between the vaccinal strain (YFV-17DD) could facilitate vaccine coverage surveillance, enabling the development of strategies to avoid new outbreaks. In this study, peptides were designed and subjected to microarray procedures with sera collected from individuals infected by WT-YFV and 17DD–YFV of YFV during the Brazilian outbreak of YFV in 2017/2018. From 222 screened peptides, around ten could potentially integrate serological approaches aiming to differentiate vaccinated individuals from naturally infected individuals. Among those peptides, one was synthesized and validated through ELISA.
Collapse
|
28
|
Celone M, Pecor DB, Potter A, Richardson A, Dunford J, Pollett S. An ecological niche model to predict the geographic distribution of Haemagogus janthinomys, Dyar, 1921 a yellow fever and Mayaro virus vector, in South America. PLoS Negl Trop Dis 2022; 16:e0010564. [PMID: 35802748 PMCID: PMC9299311 DOI: 10.1371/journal.pntd.0010564] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/20/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Yellow fever virus (YFV) has a long history of impacting human health in South America. Mayaro virus (MAYV) is an emerging arbovirus of public health concern in the Neotropics and its full impact is yet unknown. Both YFV and MAYV are primarily maintained via a sylvatic transmission cycle but can be opportunistically transmitted to humans by the bites of infected forest dwelling Haemagogus janthinomys Dyar, 1921. To better understand the potential risk of YFV and MAYV transmission to humans, a more detailed understanding of this vector species’ distribution is critical. This study compiled a comprehensive database of 177 unique Hg. janthinomys collection sites retrieved from the published literature, digitized museum specimens and publicly accessible mosquito surveillance data. Covariate analysis was performed to optimize a selection of environmental (topographic and bioclimatic) variables associated with predicting habitat suitability, and species distributions modelled across South America using a maximum entropy (MaxEnt) approach. Our results indicate that suitable habitat for Hg. janthinomys can be found across forested regions of South America including the Atlantic forests and interior Amazon. Mayaro virus is a neglected tropical disease and there is insufficient evidence to define its geographic range. The mosquito Haemagogus janthinomys is a primary vector of Mayaro and its distribution is largely unknown at a sub-country scale. Building compendiums of collection data and creating ecological niche models provides a more precise estimation of vector species potential habitat. Our dataset stands as one of the most expansive existing for collection data of this species combining data published in literature, publicly available data repositories and digitized museum specimen records. Comparing results of niche models with near real time environmental data can give even better predictions of areas where Mayaro virus exposure could occur. The methods and results of this study can be replicated for any disease/vector of interest so long as there is data discoverable through the scientific literature, public repositories, or other civilian and governmental agencies willing to share.
Collapse
Affiliation(s)
- Michael Celone
- Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - David Brooks Pecor
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Walter Reed Biosystematics Unit, Suitland, Maryland, United States of America
- * E-mail:
| | - Alexander Potter
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Walter Reed Biosystematics Unit, Suitland, Maryland, United States of America
| | - Alec Richardson
- Department of Entomology, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- Walter Reed Biosystematics Unit, Suitland, Maryland, United States of America
| | - James Dunford
- Uniformed Services University of Health Sciences, Bethesda, Maryland, United States of America
| | - Simon Pollett
- Infectious Disease Clinical Research Program, Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, United States of America
| |
Collapse
|
29
|
Silva TMRD, Nogueira de Sá ACMG, Prates EJS, Rodrigues DE, Silva TPRD, Matozinhos FP, Vieira EWR. Yellow fever vaccination before and during the covid-19 pandemic in Brazil. Rev Saude Publica 2022; 56:45. [PMID: 35703600 PMCID: PMC9165638 DOI: 10.11606/s1518-8787.2022056004503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To analyze the number of yellow fever vaccine doses administered before and during the covid-19 pandemic in Brazil. METHODS This is an ecological, time series study based on data from the National Immunization Program. Differences between the median number of yellow fever vaccine doses administered in Brazil and in its regions before (from April/2019 to March/2020) and after (from April/2020 to March/2021) the implementation of social distancing measures in the country were assessed via the Mann-Whitney test. Prais-Winsten regression models were used for time series analyses. RESULTS We found a reduction in the median number of yellow fever vaccine doses administered in Brazil and in its regions: North (-34.71%), Midwest (-21.72%), South (-63.50%), and Southeast (-34.42%) (p < 0.05). Series showed stationary behavior in Brazil and in its five regions during the covid-19 pandemic (p > 0.05). Brazilian states also showed stationary trends, except for two states which recorded an increasing trend in the number of administered yellow fever vaccine doses, namely: Alagoas State (before: β = 64, p = 0.081; after: β = 897, p = 0.039), which became a yellow fever vaccine recommendation zone, and Roraima State (before: β = 68, p = 0.724; after: β = 150, p = 0.000), which intensified yellow fever vaccinations due to a yellow fever case confirmation in a Venezuelan State in 2020. CONCLUSION The reduced number of yellow fever vaccine doses administered during the covid-19 pandemic in Brazil may favor the reemergence of urban yellow fever cases in the country.
Collapse
Affiliation(s)
- Tércia Moreira Ribeiro da Silva
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | | | - Elton Junio Sady Prates
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | | | - Thales Philipe Rodrigues da Silva
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | - Fernanda Penido Matozinhos
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| | - Ed Wilson Rodrigues Vieira
- Universidade Federal de Minas Gerais. Escola de Enfermagem. Departamento de Enfermagem Materno-Infantil e Saúde Pública. Belo Horizonte, MG, Brasil
| |
Collapse
|
30
|
de Oliveira Figueiredo P, Stoffella-Dutra AG, Costa GB, de Oliveira JS, Amaral CD, Alves PA, Filho JDA, Paz GF, Tonelli GB, Kroon EG, Drumond BP, Paglia AP, de Oliveira DB, de Souza Trindade G. Absence of yellow fever virus circulation in wildlife rodents from Brazil. Braz J Microbiol 2022; 53:647-654. [PMID: 35133637 PMCID: PMC9151931 DOI: 10.1007/s42770-022-00688-3] [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: 04/27/2021] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
Yellow fever (YF), caused by the yellow fever virus (YFV), is an emerging viral zoonosis that affects humans and non-human primates (NHP). In South America, YF is naturally maintained through enzootic/sylvatic cycles involving NHPs and mosquitoes (Haemagogus and Sabethes). In this study, we retrospectively analyzed wildlife rodents to better understand their role in a potential alternative YF sylvatic cycle. The plaque reduction neutralization test was performed to detect anti-YFV antibodies, while qPCR targeting the NS5 region of flaviviruses and standard PCR targeting the CprM region were applied to detect YFV RNA in tissue and blood samples. YFV was not evidenced in any of the tested samples. These findings provide additional information regarding sylvatic YFV and emphasize the importance of YFV surveillance in wild animals as potential reservoirs/hosts given the well-established enzootic cycle in the studied areas, mainly in the Atlantic Forest.
Collapse
Affiliation(s)
- Poliana de Oliveira Figueiredo
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Ana Gabriella Stoffella-Dutra
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| | - Galileu Barbosa Costa
- Departamento de Ciências da Saúde, Universidade Estadual de Santa Cruz, Campus Soane Nazaré de Andrade, Rodovia Jorge Amado, Km 16, Salobrinho, Ilhéus, BA, 45662-900, Brazil.
| | - Jaqueline Silva de Oliveira
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Carolina Dourado Amaral
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Pedro Augusto Alves
- Grupo de Imunologia de Doenças Virais, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | | | - Gustavo Fontes Paz
- Grupo de Estudos em Leishmanioses, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | - Gabriel Barbosa Tonelli
- Grupo de Estudos em Leishmanioses, Instituto René Rachou, Fiocruz-Minas, Belo Horizonte, MG, Brazil
| | - Erna Geessien Kroon
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Betânia Paiva Drumond
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Adriano Pereira Paglia
- Laboratório de Ecologia e Conservação, Departamento de Biologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Danilo Bretas de Oliveira
- Centro Integrado de Pesquisa em Saúde, Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Brazil
| | - Giliane de Souza Trindade
- Laboratório de Vírus, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Campus Pampulha, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
31
|
Desantis J, Felicetti T, Cannalire R. An overview on small molecules acting as broad spectrum-agents for yellow fever infection. Expert Opin Drug Discov 2022; 17:755-773. [PMID: 35638299 DOI: 10.1080/17460441.2022.2084529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Yellow Fever virus (YFV) is a mosquito-borne flavivirus, endemic in 47 countries in Africa and South America, which causes febrile symptoms that can evolve in 15% of the patients to serious haemorrhagic conditions, liver injury, and multiorgan failure. Although a highly effective vaccine (YF-17D vaccine) is available, to date, no antiviral drugs have been approved for the prevention and treatment of YFV infections. AREAS COVERED This review article focuses on the description of viral targets that have been considered within YFV and flavivirus drug discovery studies and on the most relevant candidates reported so far that elicit broad-spectrum inhibition against relevant strains and mutants of YFV. EXPERT OPINION Considering the growing interest on (re)emerging vector-borne viral infections, it is expected that flavivirus drug discovery will quickly deliver potential candidates for clinical evaluation. Due to similarity among flaviviral targets, several candidates identified against different flaviviruses have shown broad-spectrum activity, thus exhibiting anti-YFV activity, as well. In this regard, it would be desirable to routinely include the assessment of antiviral activity against different YFV strains. On the other hand, the development of host targeting agents are still at an initial stage and deserve further focused efforts.
Collapse
Affiliation(s)
- Jenny Desantis
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via Elce di Sotto 8, 06123, Perugia, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo 1, 06123, Perugia, Italy
| | - Rolando Cannalire
- Department of Pharmacy, University of Napoli "Federico II", Via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
32
|
Lücke AC, vom Hemdt A, Wieseler J, Fischer C, Feldmann M, Rothenfusser S, Drexler JF, Kümmerer BM. High-Throughput Platform for Detection of Neutralizing Antibodies Using Flavivirus Reporter Replicon Particles. Viruses 2022; 14:v14020346. [PMID: 35215941 PMCID: PMC8880525 DOI: 10.3390/v14020346] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 11/16/2022] Open
Abstract
Flavivirus outbreaks require fast and reliable diagnostics that can be easily adapted to newly emerging and re-emerging flaviviruses. Due to the serological cross-reactivity among flavivirus antibodies, neutralization tests (NT) are considered the gold standard for sero-diagnostics. Here, we first established wild-type single-round infectious virus replicon particles (VRPs) by packaging a yellow fever virus (YFV) replicon expressing Gaussia luciferase (Gluc) with YFV structural proteins in trans using a double subgenomic Sindbis virus (SINV) replicon. The latter expressed the YFV envelope proteins prME via the first SINV subgenomic promoter and the capsid protein via a second subgenomic SINV promoter. VRPs were produced upon co-electroporation of replicon and packaging RNA. Introduction of single restriction enzyme sites in the packaging construct flanking the prME sequence easily allowed to exchange the prME moiety resulting in chimeric VRPs that have the surface proteins of other flaviviruses including dengue virus 1-4, Zika virus, West Nile virus, and tick-borne encephalitis virus. Besides comparing the YF-VRP based NT assay to a YF reporter virus NT assay, we analyzed the neutralization efficiencies of different human anti-flavivirus sera or a monoclonal antibody against all established VRPs. The assays were performed in a 96-well high-throughput format setting with Gluc as readout in comparison to classical plaque reduction NTs indicating that the VRP-based NT assays are suitable for high-throughput analyses of neutralizing flavivirus antibodies.
Collapse
Affiliation(s)
- Arlen-Celina Lücke
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; (A.-C.L.); (A.v.H.); (J.W.); (M.F.)
| | - Anja vom Hemdt
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; (A.-C.L.); (A.v.H.); (J.W.); (M.F.)
| | - Janett Wieseler
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; (A.-C.L.); (A.v.H.); (J.W.); (M.F.)
| | - Carlo Fischer
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universtät Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (C.F.); (J.F.D.)
| | - Marie Feldmann
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; (A.-C.L.); (A.v.H.); (J.W.); (M.F.)
| | - Simon Rothenfusser
- Division of Clinical Pharmacology, University Hospital, LMU Munich, 80337 Munich, Germany;
- Unit Clinical Pharmacology (EKliP), Helmholtz Center for Environmental Health, 80337 Munich, Germany
| | - Jan Felix Drexler
- Institute of Virology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universtät Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany; (C.F.); (J.F.D.)
- Martinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119435 Moskow, Russia
- German Center for Infection Research (DZIF), Associated Partner Site Berlin, 10117 Berlin, Germany
| | - Beate Mareike Kümmerer
- Institute of Virology, Medical Faculty, University of Bonn, 53127 Bonn, Germany; (A.-C.L.); (A.v.H.); (J.W.); (M.F.)
- German Center for Infection Research (DZIF), Associated Partner Site Bonn-Cologne, 53127 Bonn, Germany
- Correspondence:
| |
Collapse
|
33
|
Abstract
Infectious diseases emerge via many routes and may need to overcome stepwise bottlenecks to burgeon into epidemics and pandemics. About 60% of human infections have animal origins, whereas 40% either co-evolved with humans or emerged from non-zoonotic environmental sources. Although the dynamic interaction between wildlife, domestic animals, and humans is important for the surveillance of zoonotic potential, exotic origins tend to be overemphasized since many zoonoses come from anthropophilic wild species (for example, rats and bats). We examine the equivocal evidence of whether the appearance of novel infections is accelerating and relate technological developments to the risk of novel disease outbreaks. Then we briefly compare selected epidemics, ancient and modern, from the Plague of Athens to COVID-19.
Collapse
Affiliation(s)
- Robin A Weiss
- Division of Infection & Immunity, University College London, London, UK
| | - Neeraja Sankaran
- The Descartes Centre for the History and Philosophy of the Sciences and the Humanities, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
34
|
Li SL, Acosta AL, Hill SC, Brady OJ, de Almeida MAB, Cardoso JDC, Hamlet A, Mucci LF, Telles de Deus J, Iani FCM, Alexander NS, Wint GRW, Pybus OG, Kraemer MUG, Faria NR, Messina JP. Mapping environmental suitability of Haemagogus and Sabethes spp. mosquitoes to understand sylvatic transmission risk of yellow fever virus in Brazil. PLoS Negl Trop Dis 2022; 16:e0010019. [PMID: 34995277 PMCID: PMC8797211 DOI: 10.1371/journal.pntd.0010019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 01/28/2022] [Accepted: 11/23/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Yellow fever (YF) is an arboviral disease which is endemic to Brazil due to a sylvatic transmission cycle maintained by infected mosquito vectors, non-human primate (NHP) hosts, and humans. Despite the existence of an effective vaccine, recent sporadic YF epidemics have underscored concerns about sylvatic vector surveillance, as very little is known about their spatial distribution. Here, we model and map the environmental suitability of YF's main vectors in Brazil, Haemagogus spp. and Sabethes spp., and use human population and NHP data to identify locations prone to transmission and spillover risk. METHODOLOGY/PRINCIPAL FINDINGS We compiled a comprehensive set of occurrence records on Hg. janthinomys, Hg. leucocelaenus, and Sabethes spp. from 1991-2019 using primary and secondary data sources. Linking these data with selected environmental and land-cover variables, we adopted a stacked regression ensemble modelling approach (elastic-net regularized GLM, extreme gradient boosted regression trees, and random forest) to predict the environmental suitability of these species across Brazil at a 1 km x 1 km resolution. We show that while suitability for each species varies spatially, high suitability for all species was predicted in the Southeastern region where recent outbreaks have occurred. By integrating data on NHP host reservoirs and human populations, our risk maps further highlight municipalities within the region that are prone to transmission and spillover. CONCLUSIONS/SIGNIFICANCE Our maps of sylvatic vector suitability can help elucidate potential locations of sylvatic reservoirs and be used as a tool to help mitigate risk of future YF outbreaks and assist in vector surveillance. Furthermore, at-risk regions identified from our work could help disease control and elucidate gaps in vaccination coverage and NHP host surveillance.
Collapse
Affiliation(s)
- Sabrina L. Li
- School of Geography and the Environment, University of Oxford, Oxford, United Kingdom
- * E-mail: (SLL); (JPM)
| | - André L. Acosta
- Departamento de Ecologia, Instituto de Biociências, Laboratório de Ecologia de Paisagens e Conservação—LEPAC, Universidade de São Paulo, São Paulo, Brazil
| | - Sarah C. Hill
- Department of Pathobiology and Population Sciences, Royal Veterinary College London, London, United Kingdom
| | - Oliver J. Brady
- Centre for the Mathematical Modelling of Infectious Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Department of Infectious Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Marco A. B. de Almeida
- State Centre of Health Surveillance, Rio Grande do Sul State Health Secretariat, Rio Grande do Sul, Brazil
| | - Jader da C. Cardoso
- State Centre of Health Surveillance, Rio Grande do Sul State Health Secretariat, Rio Grande do Sul, Brazil
| | - Arran Hamlet
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Luis F. Mucci
- Superintendence for Endemic Diseases Control, São Paulo State Health Secretariat, São Paulo, Brazil
| | - Juliana Telles de Deus
- Superintendence for Endemic Diseases Control, São Paulo State Health Secretariat, São Paulo, Brazil
| | | | - Neil S. Alexander
- Environmental Research Group Oxford, c/o Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - G. R. William Wint
- Environmental Research Group Oxford, c/o Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Oliver G. Pybus
- Department of Pathobiology and Population Sciences, Royal Veterinary College London, London, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | - Nuno R. Faria
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Departamento de Molestias Infecciosas e Parasitarias & Instituto de Medicina Tropical da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Jane P. Messina
- School of Geography and the Environment, University of Oxford, Oxford, United Kingdom
- Oxford School of Global and Area Studies, University of Oxford, Oxford, United Kingdom
- * E-mail: (SLL); (JPM)
| |
Collapse
|
35
|
Haslwanter D, Lasso G, Wec AZ, Furtado ND, Raphael LMS, Tse AL, Sun Y, Stransky S, Pedreño-Lopez N, Correia CA, Bornholdt ZA, Sakharkar M, Avelino-Silva VI, Moyer CL, Watkins DI, Kallas EG, Sidoli S, Walker LM, Bonaldo MC, Chandran K. Genotype-specific features reduce the susceptibility of South American yellow fever virus strains to vaccine-induced antibodies. Cell Host Microbe 2022; 30:248-259.e6. [PMID: 34998466 PMCID: PMC10067022 DOI: 10.1016/j.chom.2021.12.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/01/2021] [Accepted: 12/10/2021] [Indexed: 12/13/2022]
Abstract
The resurgence of yellow fever in South America has prompted vaccination against the etiologic agent, yellow fever virus (YFV). Current vaccines are based on a live-attenuated YF-17D virus derived from a virulent African isolate. The capacity of these vaccines to induce neutralizing antibodies against the vaccine strain is used as a surrogate for protection. However, the sensitivity of genetically distinct South American strains to vaccine-induced antibodies is unknown. We show that antiviral potency of the polyclonal antibody response in vaccinees is attenuated against an emergent Brazilian strain. This reduction was attributable to amino acid changes at two sites in central domain II of the glycoprotein E, including multiple changes at the domain I-domain II hinge, which are unique to and shared among most South American YFV strains. Our findings call for a reevaluation of current approaches to YFV immunological surveillance in South America and suggest approaches for updating vaccines.
Collapse
Affiliation(s)
- Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | | | - Nathália Dias Furtado
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil
| | - Lidiane Menezes Souza Raphael
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil
| | - Alexandra L Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA
| | - Yan Sun
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Núria Pedreño-Lopez
- Department of Pathology, The George Washington University, Washington, DC 20037, USA
| | - Carolina Argondizo Correia
- Laboratório de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | | | | | - Vivian I Avelino-Silva
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | | | - David I Watkins
- Department of Pathology, The George Washington University, Washington, DC 20037, USA
| | - Esper G Kallas
- Departamento de Moléstias Infecciosas e Parasitárias, Faculdade de Medicina, Universidade de São Paulo, 01246-903 São Paulo, Brazil
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Laura M Walker
- Adimab, LLC, Lebanon, NH 03766, USA; Adagio Therapeutics Inc., Waltham, MA 02451, USA
| | - Myrna C Bonaldo
- Laboratório de Biologia Molecular de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, 21040-360 Rio de Janeiro, Brazil.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, The Bronx, NY 10461, USA.
| |
Collapse
|
36
|
van Leur SW, Heunis T, Munnur D, Sanyal S. Pathogenesis and virulence of flavivirus infections. Virulence 2021; 12:2814-2838. [PMID: 34696709 PMCID: PMC8632085 DOI: 10.1080/21505594.2021.1996059] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/06/2021] [Accepted: 10/15/2021] [Indexed: 11/01/2022] Open
Abstract
The Flavivirus genus consists of >70 members including several that are considered significant human pathogens. Flaviviruses display a broad spectrum of diseases that can be roughly categorised into two phenotypes - systemic disease involving haemorrhage exemplified by dengue and yellow Fever virus, and neurological complications associated with the likes of West Nile and Zika viruses. Attempts to develop vaccines have been variably successful against some. Besides, mosquito-borne flaviviruses can be vertically transmitted in the arthropods, enabling long term persistence and the possibility of re-emergence. Therefore, developing strategies to combat disease is imperative even if vaccines become available. The cellular interactions of flaviviruses with their human hosts are key to establishing the viral lifecycle on the one hand, and activation of host immunity on the other. The latter should ideally eradicate infection, but often leads to immunopathological and neurological consequences. In this review, we use Dengue and Zika viruses to discuss what we have learned about the cellular and molecular determinants of the viral lifecycle and the accompanying immunopathology, while highlighting current knowledge gaps which need to be addressed in future studies.
Collapse
Affiliation(s)
| | - Tiaan Heunis
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| | - Deeksha Munnur
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| | - Sumana Sanyal
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, OxfordOX1 3RE, UK
| |
Collapse
|
37
|
Berthet M, Mesbahi G, Duvot G, Zuberbühler K, Cäsar C, Bicca-Marques JC. Dramatic decline in a titi monkey population after the 2016-2018 sylvatic yellow fever outbreak in Brazil. Am J Primatol 2021; 83:e23335. [PMID: 34609763 DOI: 10.1002/ajp.23335] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 09/03/2021] [Accepted: 09/25/2021] [Indexed: 11/07/2022]
Abstract
Platyrrhini are highly vulnerable to the yellow fever (YF) virus. From 2016 to 2018, the Atlantic Forest of southeast Brazil faced its worst sylvatic YF outbreak in about a century, thought to have killed thousands of primates. It is essential to assess the impact of this epidemic on threatened primate assemblages to design effective conservation strategies. In this study, we assessed the impact of the 2016-2018 YF outbreak on a geographically isolated population of Near Threatened black-fronted titi monkeys (Callicebus nigrifrons) in two Atlantic Forest patches of the Santuário do Caraça, MG, Brazil. Extensive preoutbreak monitoring, conducted between 2008 and 2016, revealed that the home range and group sizes of the population remained stable. In 2016, the population size was estimated at 53-57 individuals in 11-12 groups. We conducted monitoring and playback surveys in 2019 and found that the population had decreased by 68% in one forest patch and completely vanished in the other, resulting in a combined decline of 80%. We discuss this severe loss of a previously stable population and conclude that it was highly likely caused by the YF outbreak. The remaining population is at risk of disappearing completely because of its small size and geographic isolation. A systematic population surveys of C. nigrifrons, along other sensible Platyrrhini species, is needed to re-evaluate their current conservation status.
Collapse
Affiliation(s)
- Mélissa Berthet
- Département d'études cognitives, Institut Jean Nicod, ENS, EHESS, CNRS, PSL Research University, Paris, France
| | - Geoffrey Mesbahi
- Université de Lorraine, INRAE, LAE, Nancy, France.,Parc Naturel Régional des Vosges du Nord, La Petite Pierre, France
| | - Guilhem Duvot
- Département d'études cognitives, Institut Jean Nicod, ENS, EHESS, CNRS, PSL Research University, Paris, France
| | - Klaus Zuberbühler
- School of Psychology & Neurosciences, University of St Andrews, Scotland, UK.,Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | | | - Júlio Cèsar Bicca-Marques
- Escola de Ciências da Saúde e da Vida, Laboratório de Primatologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
38
|
Andrade MDS, Campos FS, Campos AAS, Abreu FVS, Melo FL, Sevá ADP, Cardoso JDC, Dos Santos E, Born LC, da Silva CMD, Müller NFD, de Oliveira CH, da Silva AJJ, Simonini-Teixeira D, Bernal-Valle S, Mares-Guia MAMM, Albuquerque GR, Romano APM, Franco AC, Ribeiro BM, Roehe PM, de Almeida MAB. Real-Time Genomic Surveillance during the 2021 Re-Emergence of the Yellow Fever Virus in Rio Grande do Sul State, Brazil. Viruses 2021; 13:v13101976. [PMID: 34696408 PMCID: PMC8539658 DOI: 10.3390/v13101976] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 02/06/2023] Open
Abstract
The 2021 re-emergence of yellow fever in non-human primates in the state of Rio Grande do Sul (RS), southernmost Brazil, resulted in the death of many howler monkeys (genus Alouatta) and led the state to declare a Public Health Emergency of State Importance, despite no human cases reported. In this study, near-complete genomes of yellow fever virus (YFV) recovered from the outbreak were sequenced and examined aiming at a better understanding of the phylogenetic relationships and the spatio-temporal dynamics of the virus distribution. Our results suggest that the most likely sequence of events involved the reintroduction of YFV from the state of São Paulo to RS through the states of Paraná and Santa Catarina, by the end of 2020. These findings reinforce the role of genomic surveillance in determining the pathways of distribution of the virus and in providing references for the implementation of preventive measures for populations in high risk areas.
Collapse
Affiliation(s)
- Miguel de S. Andrade
- Baculovirus Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília 70910-900, Distrito Federal, Brazil; (M.d.S.A.); (F.L.M.); (B.M.R.)
| | - Fabrício S. Campos
- Bioinformatics and Biotechnology Laboratory, Campus of Gurupi, Federal University of Tocantins, Gurupi 77410-570, Tocantins, Brazil;
| | - Aline A. S. Campos
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
| | - Filipe V. S. Abreu
- Insect Behavior Laboratory, Federal Institute of Northern Minas Gerais, Salinas 39560-000, Minas Gerais, Brazil; (F.V.S.A.); (C.H.d.O.); (A.J.J.d.S.)
| | - Fernando L. Melo
- Baculovirus Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília 70910-900, Distrito Federal, Brazil; (M.d.S.A.); (F.L.M.); (B.M.R.)
| | - Anaiá da P. Sevá
- Department of Agricultural and Environmental Sciences, Santa Cruz State University, Ilhéus 45662-900, Bahia, Brazil; (A.d.P.S.); (D.S.-T.); (S.B.-V.); (G.R.A.)
| | - Jader da C. Cardoso
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
| | - Edmilson Dos Santos
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
| | - Lucas C. Born
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
| | - Cláudia M. D. da Silva
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
| | - Nicolas F. D. Müller
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Rio Grande do Sul, Brazil; (N.F.D.M.); (A.C.F.); (P.M.R.)
| | - Cirilo H. de Oliveira
- Insect Behavior Laboratory, Federal Institute of Northern Minas Gerais, Salinas 39560-000, Minas Gerais, Brazil; (F.V.S.A.); (C.H.d.O.); (A.J.J.d.S.)
| | - Alex J. J. da Silva
- Insect Behavior Laboratory, Federal Institute of Northern Minas Gerais, Salinas 39560-000, Minas Gerais, Brazil; (F.V.S.A.); (C.H.d.O.); (A.J.J.d.S.)
| | - Danilo Simonini-Teixeira
- Department of Agricultural and Environmental Sciences, Santa Cruz State University, Ilhéus 45662-900, Bahia, Brazil; (A.d.P.S.); (D.S.-T.); (S.B.-V.); (G.R.A.)
| | - Sofía Bernal-Valle
- Department of Agricultural and Environmental Sciences, Santa Cruz State University, Ilhéus 45662-900, Bahia, Brazil; (A.d.P.S.); (D.S.-T.); (S.B.-V.); (G.R.A.)
| | - Maria A. M. M. Mares-Guia
- Flavivirus Laboratory, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Rio de Janeiro, Brazil;
| | - George R. Albuquerque
- Department of Agricultural and Environmental Sciences, Santa Cruz State University, Ilhéus 45662-900, Bahia, Brazil; (A.d.P.S.); (D.S.-T.); (S.B.-V.); (G.R.A.)
| | - Alessandro P. M. Romano
- General Coordination of Arbovirus Surveillance, Ministry of Health, Brasília 70058-900, Distrito Federal, Brazil;
| | - Ana C. Franco
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Rio Grande do Sul, Brazil; (N.F.D.M.); (A.C.F.); (P.M.R.)
| | - Bergmann M. Ribeiro
- Baculovirus Laboratory, Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, Brasília 70910-900, Distrito Federal, Brazil; (M.d.S.A.); (F.L.M.); (B.M.R.)
| | - Paulo M. Roehe
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre 90050-170, Rio Grande do Sul, Brazil; (N.F.D.M.); (A.C.F.); (P.M.R.)
| | - Marco A. B. de Almeida
- State Center of Health Surveillance, Rio Grande do Sul State Health Department, Porto Alegre 90610-000, Rio Grande do Sul, Brazil; (A.A.S.C.); (J.d.C.C.); (E.d.S.); (L.C.B.); (C.M.D.d.S.)
- Correspondence:
| |
Collapse
|
39
|
Viral and Prion Infections Associated with Central Nervous System Syndromes in Brazil. Viruses 2021; 13:v13071370. [PMID: 34372576 PMCID: PMC8310075 DOI: 10.3390/v13071370] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 12/12/2022] Open
Abstract
Virus-induced infections of the central nervous system (CNS) are among the most serious problems in public health and can be associated with high rates of morbidity and mortality, mainly in low- and middle-income countries, where these manifestations have been neglected. Typically, herpes simplex virus 1 and 2, varicella-zoster, and enterovirus are responsible for a high number of cases in immunocompetent hosts, whereas other herpesviruses (for example, cytomegalovirus) are the most common in immunocompromised individuals. Arboviruses have also been associated with outbreaks with a high burden of neurological disorders, such as the Zika virus epidemic in Brazil. There is a current lack of understanding in Brazil about the most common viruses involved in CNS infections. In this review, we briefly summarize the most recent studies and findings associated with the CNS, in addition to epidemiological data that provide extensive information on the circulation and diversity of the most common neuro-invasive viruses in Brazil. We also highlight important aspects of the prion-associated diseases. This review provides readers with better knowledge of virus-associated CNS infections. A deeper understanding of these infections will support the improvement of the current surveillance strategies to allow the timely monitoring of the emergence/re-emergence of neurotropic viruses.
Collapse
|
40
|
Yan L, Song Y, Xia K, He P, Zhang F, Chen S, Pouliot R, Weiss DJ, Tandon R, Bates JT, Ederer DR, Mitra D, Sharma P, Davis A, Linhardt RJ. Heparan sulfates from bat and human lung and their binding to the spike protein of SARS-CoV-2 virus. Carbohydr Polym 2021; 260:117797. [PMID: 33712145 PMCID: PMC7882221 DOI: 10.1016/j.carbpol.2021.117797] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/22/2021] [Accepted: 02/06/2021] [Indexed: 12/27/2022]
Abstract
Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) has resulted in a pandemic and continues to spread at an unprecedented rate around the world. Although a vaccine has recently been approved, there are currently few effective therapeutics to fight its associated disease in humans, COVID-19. SARS-CoV-2 and the related severe acute respiratory syndrome (SARS-CoV-1), and Middle East respiratory syndrome (MERS-CoV) result from zoonotic respiratory viruses that have bats as the primary host and an as yet unknown secondary host. While each of these viruses has different protein-based cell-surface receptors, each rely on the glycosaminoglycan, heparan sulfate as a co-receptor. In this study we compare, for the first time, differences and similarities in the structure of heparan sulfate in human and bat lungs. Furthermore, we show that the spike glycoprotein of COVID-19 binds 3.5 times stronger to human lung heparan sulfate than bat lung heparan sulfate.
Collapse
Affiliation(s)
- Lufeng Yan
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, 310058, China; Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States
| | - Yuefan Song
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States
| | - Ke Xia
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States
| | - Peng He
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, 310058, China
| | - Robert Pouliot
- Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Daniel J Weiss
- Larner College of Medicine, University of Vermont, Burlington, VT, United States
| | - Ritesh Tandon
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, United States
| | - John T Bates
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, United States
| | - Dallas R Ederer
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, United States
| | - Dipanwita Mitra
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, United States
| | - Poonam Sharma
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS, 39216, United States
| | - April Davis
- Rabies Laboratory, New York State Department of Health Wadsworth Center, Albany, New York, United States
| | - Robert J Linhardt
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States; Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, 12180, United States.
| |
Collapse
|
41
|
Dengue-2 and Guadeloupe Mosquito Virus RNA Detected in Aedes ( Stegomyia) spp. Collected in a Vehicle Impound Yard in Santo André, SP, Brazil. INSECTS 2021; 12:insects12030248. [PMID: 33809477 PMCID: PMC8001461 DOI: 10.3390/insects12030248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/17/2022]
Abstract
In 2018-2019, we conducted mosquito collections in a municipal vehicle impound yard, which is 10 km from the Serra do Mar Environmental Protection Area in Santo André, SP, Brazil. Our aim is to study arboviruses in the impound yard, to understand the transmission of arboviruses in an urban environment in Brazil. We captured the mosquitoes using human-landing catches and processed them for arbovirus detection by conventional and quantitative RT-PCR assays. We captured two mosquito species, Aedes aegypti (73 total specimens; 18 females and 55 males) and Ae. albopictus (34 specimens; 27 females and 7 males). The minimum infection rate for DENV-2 was 11.5 per 1000 (CI95%: 1-33.9). The detection of DENV-2 RNA in an Ae. albopictus female suggests that this virus might occur in high infection rates in the sampled mosquito population and is endemic in the urban areas of Santo André. In addition, Guadeloupe mosquito virus RNA was detected in an Ae. aegypti female. To our knowledge, this was the first detection of the Guadeloupe mosquito virus in Brazil.
Collapse
|
42
|
Special Issue "Emerging Viruses 2020: Surveillance, Prevention, Evolution and Control". Viruses 2021; 13:v13020251. [PMID: 33562062 PMCID: PMC7915717 DOI: 10.3390/v13020251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/25/2022] Open
|