51
|
Fialho EMS, Veras EM, Jesus CMD, Gomes LN, Khouri R, Sousa PS, Ribeiro MRC, Batista RFL, Costa LC, Nascimento FRF, Silva AAM, Soeiro-Pereira PV. Maternal Th17 Profile after Zika Virus Infection Is Involved in Congenital Zika Syndrome Development in Children. Viruses 2023; 15:1320. [PMID: 37376620 DOI: 10.3390/v15061320] [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: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Brazil is one of the countries that experienced an epidemic of microcephaly and other congenital manifestations related to maternal Zika virus infection which can result in Congenital Zika Syndrome (CZS). Since the Zika virus can modulate the immune system, studying mothers' and children's immune profiles become essential to better understanding CZS development. Therefore, we investigated the lymphocyte population profile of children who developed CZS and their mothers' immune response in this study. The study groups were formed from the Plaque Reduction Neutralization Test (PRNT) (CZS+ group) result. To evaluate the lymphocyte population profile, we performed phenotyping of peripheral lymphocytes and quantification of serum cytokine levels. The immunophenotyping and cytokine profile was correlated between CSZ+ children and their mothers. Both groups exhibited increased interleukin-17 levels and a reduction in the subpopulation of CD4+ T lymphocytes. In contrast, the maternal group showed a reduction in the population of B lymphocytes. Thus, the development of CZS is related to the presence of an inflammatory immune profile in children and their mothers characterized by Th17 activation.
Collapse
Affiliation(s)
- Eder M S Fialho
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Emanoel M Veras
- Medical School, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Caroline M de Jesus
- Health and Technology Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
| | - Líllian N Gomes
- Department of Immunology, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Ricardo Khouri
- Gonçalo Moniz Research Institute, FIOCRUZ-Bahia, Salvador 40296-710, BA, Brazil
| | - Patrícia S Sousa
- Reference Center on Neurodevelopment, Assistance and Rehabilitation of Children/NINAR-State Department of Health of the State of Maranhão, São Luís 65077-357, MA, Brazil
| | - Marizélia R C Ribeiro
- Department of Medicine III, Federal University of Maranhão, São Luís 65020-240, MA, Brazil
| | - Rosângela F L Batista
- Department of Public Health, Federal University of Maranhão, São Luís 65020-060, MA, Brazil
| | - Luciana C Costa
- Department of Public Health, Federal University of Maranhão, São Luís 65020-060, MA, Brazil
| | - Flávia R F Nascimento
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
- Department of Pathology, Federal University of Maranhão, São Luís 65065-545, MA, Brazil
| | - Antônio A M Silva
- Department of Public Health, Federal University of Maranhão, São Luís 65020-060, MA, Brazil
| | - Paulo V Soeiro-Pereira
- Health Sciences Graduate Program, Biological and Health Sciences Center, Federal University of Maranhão, São Luís 65080-805, MA, Brazil
- Department of Pathology, Federal University of Maranhão, São Luís 65065-545, MA, Brazil
| |
Collapse
|
52
|
Hale GL. Flaviviruses and the Traveler: Around the World and to Your Stage. A Review of West Nile, Yellow Fever, Dengue, and Zika Viruses for the Practicing Pathologist. Mod Pathol 2023; 36:100188. [PMID: 37059228 DOI: 10.1016/j.modpat.2023.100188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023]
Abstract
Flaviviruses are a genus of single-stranded RNA viruses that impose an important and growing burden to human health. There are over 3 billion individuals living in areas where flaviviruses are endemic. Flaviviruses and their arthropod vectors (which include mosquitoes and ticks) take advantage of global travel to expand their distribution and cause severe disease in humans, and they can be grouped according to their vector and pathogenicity. The mosquito-borne flaviviruses cause a spectrum of diseases from encephalitis to hepatitis and vascular shock syndrome, congenital abnormalities, and fetal death. Neurotropic infections such as Zika virus and West Nile virus cross the blood-brain barrier and infect neurons and other cells, leading to meningoencephalitis. In the hemorrhagic fever clade, there are yellow fever virus, the prototypical hemorrhagic fever virus that infects hepatocytes, and dengue virus, which infects cells of the reticuloendothelial system and can lead to a dramatic plasma cell leakage and shock syndrome. Zika virus also causes congenital infections and fetal death and is the first and only example of a teratogenic arbovirus in humans. Diagnostic testing for flaviviruses broadly includes the detection of viral RNA in serum (particularly within the first 10 days of symptoms), viral isolation by cell culture (rarely performed due to complexity and biosafety concerns), and histopathologic evaluation with immunohistochemistry and molecular testing on formalin-fixed paraffin-embedded tissue blocks. This review focuses on 4 mosquito-borne flaviviruses-West Nile, yellow fever, dengue, and Zika virus-and discusses the mechanisms of transmission, the role of travel in geographic distribution and epidemic emergence, and the clinical and histopathologic features of each. Finally, prevention strategies such as vector control and vaccination are discussed.
Collapse
Affiliation(s)
- Gillian L Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
53
|
Coffey CH, Casper LM, Reno EM, Casper SJ, Hillis E, Klein DA, Schlein SM, Keyes LE. First-Trimester Pregnancy: Considerations for Wilderness and Remote Travel. Wilderness Environ Med 2023; 34:201-210. [PMID: 36842861 DOI: 10.1016/j.wem.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 02/28/2023]
Abstract
Women increasingly participate in outdoor activities in wilderness and remote environments. We performed a literature review to address diagnostic and therapeutic considerations during first-trimester pregnancy for remote multiday travel. Pretrip planning for pregnant patients traveling outside access to advanced medical care should include performing a transvaginal ultrasound to confirm pregnancy location and checking D rhesus status. We discuss the risk of potential travel-related infections and recommended vaccinations prior to departure based on destination. Immediate evacuation to definitive medical care is required for patients with a pregnancy of unknown location and vaginal bleeding. We propose algorithms for determining the need for evacuation and present therapeutic options for nausea and vomiting, urinary tract infections, and candidiasis in the field.
Collapse
Affiliation(s)
- Christanne H Coffey
- Department of Emergency Medicine, University of California, San Diego, San Diego, CA.
| | | | - Elaine M Reno
- Department of Emergency Medicine, University of Colorado, Aurora, CO
| | - Sierra J Casper
- Mammoth Hospital, San Diego Fire-Rescue Department, San Diego, CA
| | | | - David A Klein
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Diego, San Diego, CA
| | - Sarah M Schlein
- Larner College of Medicine, University of Vermont, Burlington, VT
| | - Linda E Keyes
- Department of Emergency Medicine, University of Colorado, Aurora, CO
| |
Collapse
|
54
|
Baltar JMC, Pavan MG, Corrêa-Antônio J, Couto-Lima D, Maciel-de-Freitas R, David MR. Gut Bacterial Diversity of Field and Laboratory-Reared Aedes albopictus Populations of Rio de Janeiro, Brazil. Viruses 2023; 15:1309. [PMID: 37376609 DOI: 10.3390/v15061309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The mosquito microbiota impacts different parameters in host biology, such as development, metabolism, immune response and vector competence to pathogens. As the environment is an important source of acquisition of host associate microbes, we described the microbiota and the vector competence to Zika virus (ZIKV) of Aedes albopictus from three areas with distinct landscapes. METHODS Adult females were collected during two different seasons, while eggs were used to rear F1 colonies. Midgut bacterial communities were described in field and F1 mosquitoes as well as in insects from a laboratory colony (>30 generations, LAB) using 16S rRNA gene sequencing. F1 mosquitoes were infected with ZIKV to determine virus infection rates (IRs) and dissemination rates (DRs). Collection season significantly affected the bacterial microbiota diversity and composition, e.g., diversity levels decreased from the wet to the dry season. Field-collected and LAB mosquitoes' microbiota had similar diversity levels, which were higher compared to F1 mosquitoes. However, the gut microbiota composition of field mosquitoes was distinct from that of laboratory-reared mosquitoes (LAB and F1), regardless of the collection season and location. A possible negative correlation was detected between Acetobacteraceae and Wolbachia, with the former dominating the gut microbiota of F1 Ae. albopictus, while the latter was absent/undetectable. Furthermore, we detected significant differences in infection and dissemination rates (but not in the viral load) between the mosquito populations, but it does not seem to be related to gut microbiota composition, as it was similar between F1 mosquitoes regardless of their population. CONCLUSIONS Our results indicate that the environment and the collection season play a significant role in shaping mosquitoes' bacterial microbiota.
Collapse
Affiliation(s)
- João M C Baltar
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Márcio G Pavan
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Jessica Corrêa-Antônio
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Dinair Couto-Lima
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| | - Rafael Maciel-de-Freitas
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
- Department of Arbovirology, Bernhard Nocht Institute of Tropical Medicine, 20359 Hamburg, Germany
| | - Mariana R David
- Laboratório de Mosquitos Transmissores de Hematozoários, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, RJ, Brazil
| |
Collapse
|
55
|
Lu AY, Gustin A, Newhouse D, Gale M. Viral Protein Accumulation of Zika Virus Variants Links with Regulation of Innate Immunity for Differential Control of Viral Replication, Spread, and Response to Interferon. J Virol 2023; 97:e0198222. [PMID: 37162358 PMCID: PMC10231147 DOI: 10.1128/jvi.01982-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/11/2023] Open
Abstract
Asian lineage Zika virus (ZIKV) strains emerged globally, causing outbreaks linked with critical clinical disease outcomes unless the virus is effectively restricted by host immunity. We have previously shown that retinoic acid-inducible gene-I (RIG-I) senses ZIKV to trigger innate immunity to direct interferon (IFN) production and antiviral responses that can control ZIKV infection. However, ZIKV proteins have been demonstrated to antagonize IFN. Here, we conducted in vitro analyses to assess how divergent prototypic ZIKV variants differ in virologic properties, innate immune regulation, and infection outcome. We comparatively assessed African lineage ZIKV/Dakar/1984/ArD41519 (ZIKV/Dakar) and Asian lineage ZIKV/Malaysia/1966/P6740 (ZIKV/Malaysia) in a human epithelial cell infection model. De novo viral sequence determination identified amino acid changes within the ZIKV/Dakar genome compared to ZIKV/Malaysia. Viral growth analyses revealed that ZIKV/Malaysia accumulated viral proteins and genome copies earlier and to higher levels than ZIKV/Dakar. Both ZIKV strains activated RIG-I/IFN regulatory factor (IRF3) and NF-κB pathways to induce inflammatory cytokine expression and types I and III IFNs. However, ZIKV/Malaysia, but not ZIKV/Dakar, potently blocked downstream IFN signaling. Remarkably, ZIKV/Dakar protein accumulation and genome replication were rescued in RIG-I knockout (KO) cells late in acute infection, resulting in ZIKV/Dakar-mediated blockade of IFN signaling. We found that RIG-I signaling specifically restricts viral protein accumulation late in acute infection where early accumulation of viral proteins in infected cells confers enhanced ability to limit IFN signaling, promoting viral replication and spread. Our results demonstrate that RIG-I-mediated innate immune signaling imparts restriction of ZIKV protein accumulation, which permits IFN signaling and antiviral actions controlling ZIKV infection. IMPORTANCE ZIKV isolates are classified under African or Asian lineages. Infection with emerging Asian lineage-derived ZIKV strains is associated with increased incidence of neurological symptoms that were not previously reported during infection with African or preemergent Asian lineage viruses. In this study, we utilized in vitro models to compare the virologic properties of and innate immune responses to two prototypic ZIKV strains from distinct lineages: African lineage ZIKV/Dakar and Asian lineage ZIKV/Malaysia. Compared to ZIKV/Dakar, ZIKV/Malaysia accumulates viral proteins earlier, replicates to higher levels, and robustly blocks IFN signaling during acute infection. Early accumulation of ZIKV/Malaysia NS5 protein confers enhanced ability to antagonize IFN signaling, dampening innate immune responses to promote viral spread. Our data identify the kinetics of viral protein accumulation as a major regulator of host innate immunity, influencing host-mediated control of ZIKV replication and spread. Importantly, these findings provide a novel framework for evaluating the virulence of emerging variants.
Collapse
Affiliation(s)
- Amy Y. Lu
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Andrew Gustin
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Daniel Newhouse
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
56
|
de Moraes L, Portilho MM, Vrancken B, Van den Broeck F, Santos LA, Cucco M, Tauro LB, Kikuti M, Silva MMO, Campos GS, Reis MG, Barral A, Barral-Netto M, Boaventura VS, Vandamme AM, Theys K, Lemey P, Ribeiro GS, Khouri R. Analyses of Early ZIKV Genomes Are Consistent with Viral Spread from Northeast Brazil to the Americas. Viruses 2023; 15:1236. [PMID: 37376536 DOI: 10.3390/v15061236] [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: 04/13/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The Americas, particularly Brazil, were greatly impacted by the widespread Zika virus (ZIKV) outbreak in 2015 and 2016. Efforts were made to implement genomic surveillance of ZIKV as part of the public health responses. The accuracy of spatiotemporal reconstructions of the epidemic spread relies on the unbiased sampling of the transmission process. In the early stages of the outbreak, we recruited patients exhibiting clinical symptoms of arbovirus-like infection from Salvador and Campo Formoso, Bahia, in Northeast Brazil. Between May 2015 and June 2016, we identified 21 cases of acute ZIKV infection and subsequently recovered 14 near full-length sequences using the amplicon tiling multiplex approach with nanopore sequencing. We performed a time-calibrated discrete phylogeographic analysis to trace the spread and migration history of the ZIKV. Our phylogenetic analysis supports a consistent relationship between ZIKV migration from Northeast to Southeast Brazil and its subsequent dissemination beyond Brazil. Additionally, our analysis provides insights into the migration of ZIKV from Brazil to Haiti and the role Brazil played in the spread of ZIKV to other countries, such as Singapore, the USA, and the Dominican Republic. The data generated by this study enhances our understanding of ZIKV dynamics and supports the existing knowledge, which can aid in future surveillance efforts against the virus.
Collapse
Affiliation(s)
- Laise de Moraes
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40026-010, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Moyra M Portilho
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Bram Vrancken
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
- Spatial Epidemiology Lab (SpELL), Université Libre de Bruxelles, 1050 Bruxelles, Belgium
| | - Frederik Van den Broeck
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
- Department of Biomedical Sciences, Antwerp Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Luciane Amorim Santos
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40026-010, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Escola Bahiana de Medicina e Saúde Pública, Salvador 41150-100, Brazil
| | - Marina Cucco
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40026-010, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Laura B Tauro
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Instituto de Biología Subtropical, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Misiones, Puerto Iguazú N3370, Argentina
| | - Mariana Kikuti
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Monaise M O Silva
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Gúbio S Campos
- Laboratório de Virologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador 40231-300, Brazil
| | - Mitermayer G Reis
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Departamento de Patologia e Medicina Legal, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40110-100, Brazil
| | - Aldina Barral
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Manoel Barral-Netto
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
| | - Viviane Sampaio Boaventura
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Hospital Santa Izabel, Salvador 40050-410, Brazil
| | - Anne-Mieke Vandamme
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
- Center for Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, 1349-008 Lisbon, Portugal
| | - Kristof Theys
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
| | - Philippe Lemey
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
| | - Guilherme S Ribeiro
- Laboratório de Patologia e Biologia Molecular, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Departamento de Medicina Preventiva e Social, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40110-100, Brazil
| | - Ricardo Khouri
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina da Bahia, Universidade Federal da Bahia, Salvador 40026-010, Brazil
- Laboratório de Enfermidades Infecciosas Transmitidas por Vetores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador 40296-710, Brazil
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
57
|
Ayusso GM, Lima MLD, da Silva Sanches PR, Santos IA, Martins DOS, da Conceição PJP, Carvalho T, da Costa VG, Bittar C, Merits A, Santos-Filho NA, Cilli EM, Jardim ACG, de Freitas Calmon M, Rahal P. The Dimeric Peptide (KKYRYHLKPF) 2K Shows Broad-Spectrum Antiviral Activity by Inhibiting Different Steps of Chikungunya and Zika Virus Infection. Viruses 2023; 15:v15051168. [PMID: 37243254 DOI: 10.3390/v15051168] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Chikungunya virus (CHIKV) and Zika virus (ZIKV) are important disease-causing agents worldwide. Currently, there are no antiviral drugs or vaccines approved to treat these viruses. However, peptides have shown great potential for new drug development. A recent study described (p-BthTX-I)2K [(KKYRYHLKPF)2K], a peptide derived from the Bothropstoxin-I toxin in the venom of the Bothrops jararacussu snake, showed antiviral activity against SARS-CoV-2. In this study, we assessed the activity of this peptide against CHIKV and ZIKV and its antiviral action in the different stages of the viral replication cycle in vitro. We observed that (p-BthTX-I)2K impaired CHIKV infection by interfering with the early steps of the viral replication cycle, reducing CHIKV entry into BHK-21 cells specifically by reducing both the attachment and internalization steps. (p-BthTX-I)2K also inhibited the ZIKV replicative cycle in Vero cells. The peptide protected the cells against ZIKV infection and decreased the levels of the viral RNA and the NS3 protein of this virus at viral post-entry steps. In conclusion, this study highlights the potential of the (p-BthTX-I)2K peptide to be a novel broad-spectrum antiviral candidate that targets different steps of the replication cycle of both CHIKV and ZIKV.
Collapse
Affiliation(s)
- Gabriela Miranda Ayusso
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| | - Maria Letícia Duarte Lima
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| | | | - Igor Andrade Santos
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil
| | - Daniel Oliveira Silva Martins
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil
| | | | - Tamara Carvalho
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| | - Vivaldo Gomes da Costa
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| | - Cíntia Bittar
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Andres Merits
- Institute of Technology, University of Tartu, 50090 Tartu, Estonia
| | | | - Eduardo Maffud Cilli
- Institute of Chemistry, São Paulo State University, Araraquara 14800-060, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil
| | - Marilia de Freitas Calmon
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| | - Paula Rahal
- Institute of Biosciences, Letters and Exact Sciences, São Paulo State University, São José do Rio Preto 15054-000, SP, Brazil
| |
Collapse
|
58
|
Nakase T, Giovanetti M, Obolski U, Lourenço J. Global transmission suitability maps for dengue virus transmitted by Aedes aegypti from 1981 to 2019. Sci Data 2023; 10:275. [PMID: 37173303 PMCID: PMC10182074 DOI: 10.1038/s41597-023-02170-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Mosquito-borne viruses increasingly threaten human populations due to accelerating changes in climate, human and mosquito migration, and land use practices. Over the last three decades, the global distribution of dengue has rapidly expanded, causing detrimental health and economic problems in many areas of the world. To develop effective disease control measures and plan for future epidemics, there is an urgent need to map the current and future transmission potential of dengue across both endemic and emerging areas. Expanding and applying Index P, a previously developed mosquito-borne viral suitability measure, we map the global climate-driven transmission potential of dengue virus transmitted by Aedes aegypti mosquitoes from 1981 to 2019. This database of dengue transmission suitability maps and an R package for Index P estimations are offered to the public health community as resources towards the identification of past, current and future transmission hotspots. These resources and the studies they facilitate can contribute to the planning of disease control and prevention strategies, especially in areas where surveillance is unreliable or non-existent.
Collapse
Affiliation(s)
- Taishi Nakase
- Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK.
| | - Marta Giovanetti
- Laboratório de Flavivírus, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, 21040-360, Brazil
- Department of Science and Technology for Humans and the Environment, University of Campus Bio-Medico di Roma, Rome, 00128, Italy
| | - Uri Obolski
- School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
- Porter School of the Environment and Earth Sciences, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
| | - José Lourenço
- Biosystems and Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, 1749-016, Portugal.
| |
Collapse
|
59
|
Wanzeller ALM, da Silva FS, Hernández LHA, Barros LJL, Freitas MNO, Santos MM, Gonçalves EDJ, Pantoja JAS, Lima CDS, Lima MF, Costa LRO, das Chagas LL, Silva IF, da Cunha TCADS, do Nascimento BLS, Vasconcelos HB, da Rosa EST, Rodrigues SG, Azevedo RDSDS, Martins LC, Casseb LMN, Chiang JO, Nunes Neto JP, Cruz ACR, Carvalho VL, Vasconcelos PFDC, da Silva EVP. Isolation of Flaviviruses and Alphaviruses with Encephalitogenic Potential Diagnosed by Evandro Chagas Institute (Pará, Brazil) in the Period of 1954-2022: Six Decades of Discoveries. Viruses 2023; 15:v15040935. [PMID: 37112917 PMCID: PMC10146763 DOI: 10.3390/v15040935] [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: 02/09/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 04/29/2023] Open
Abstract
Viruses with encephalitogenic potential can cause neurological conditions of clinical and epidemiological importance, such as Saint Louis encephalitis virus, Venezuelan equine encephalitis virus, Eastern equine encephalitis virus, Western equine encephalitis virus, Dengue virus, Zika virus, Chikungunya virus, Mayaro virus and West Nile virus. The objective of the present study was to determine the number of arboviruses with neuroinvasive potential isolated in Brazil that corresponds to the collection of viral samples belonging to the Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute (SAARB/IEC) of the Laboratory Network of National Reference for Arbovirus Diagnosis from 1954 to 2022. In the analyzed period, a total of 1,347 arbovirus samples with encephalitogenic potential were isolated from mice; 5,065 human samples were isolated exclusively by cell culture; and 676 viruses were isolated from mosquitoes. The emergence of new arboviruses may be responsible for diseases still unknown to humans, making the Amazon region a hotspot for infectious diseases due to its fauna and flora species characteristics. The detection of circulating arboviruses with the potential to cause neuroinvasive diseases is constant, which justifies the continuation of active epidemiological surveillance work that offers adequate support to the public health system regarding the virological diagnosis of circulating arboviruses in Brazil.
Collapse
Affiliation(s)
- Ana Lucia Monteiro Wanzeller
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Fabio Silva da Silva
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Leonardo Henrique Almeida Hernández
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Landerson Junior Leopoldino Barros
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Maria Nazaré Oliveira Freitas
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Maissa Maia Santos
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Ercília de Jesus Gonçalves
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Jamilla Augusta Sousa Pantoja
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Creuza de Sousa Lima
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Maxwell Furtado Lima
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Luiz Roberto Oliveira Costa
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Liliane Leal das Chagas
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Iveraldo Ferreira Silva
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Tania Cristina Alves da Silveira da Cunha
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Bruna Lais Sena do Nascimento
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Helena Baldez Vasconcelos
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Elizabeth Salbe Travassos da Rosa
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Sueli Guerreiro Rodrigues
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Raimunda do Socorro da Silva Azevedo
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Lívia Carício Martins
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Lívia Medeiros Neves Casseb
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Jannifer Oliveira Chiang
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Joaquim Pinto Nunes Neto
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Ana Cecília Ribeiro Cruz
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Valéria Lima Carvalho
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| | - Eliana Vieira Pinto da Silva
- Viral Isolation Laboratory, Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Secretariat of Health and Environment Surveillance, Ministry of Health, Ananindeua 67030-000, Brazil
| |
Collapse
|
60
|
Zhang Y, Wang Q, Zhu Z, Wang S, Tu S, Zhang Y, Zou Y, Liu Y, Liu C, Ren W, Zheng D, Zhao Y, Hu Y, Li L, Shi C, Ge S, Lin P, Xu F, Ma J, Wu X, Ma H, Wang Z, Bao J. Tracing the Origin of Genotype II African Swine Fever Virus in China by Genomic Epidemiology Analysis. Transbound Emerg Dis 2023. [DOI: 10.1155/2023/4820809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
The pandemic spread of African swine fever (ASF) has caused serious effects on the global pig industry. Virus genome sequencing and genomic epidemiology analysis play an important role in tracking the outbreaks of the disease and tracing the transmission of the virus. Here we obtained the full-length genome sequence of African swine fever virus (ASFV) in the first outbreak of ASF in China on August 3rd, 2018 and compared it with other published genotype II ASFV genomes including 9 genomes collected in China from September 2018 to October 2020. Phylogenetic analysis on genomic sequences revealed that genotype II ASFV has evolved into different genetic clusters with temporal and spatial correlation since being introduced into Europe and then Asia. There was a strong support for the monophyletic grouping of all the ASFV genome sequences from China and other Asian countries, which shared a common ancestor with those from the Central or Eastern Europe. An evolutionary rate of 1.312 × 10−5 nucleotide substitutions per site per year was estimated for genotype II ASFV genomes. Eight single nucleotide variations which located in MGF110-1L, MGF110-7L, MGF360-10L, MGF505-5R, MGF505-9R, K145R, NP419L, and I267L were identified as anchor mutations that defined genetic clusters of genotype II ASFV in Europe and Asia. This study expanded our knowledge of the molecular epidemiology of ASFV and provided valuable information for effective control of the disease.
Collapse
|
61
|
Castro-Amarante MFD, Pereira SS, Pereira LR, Santos LS, Venceslau-Carvalho AA, Martins EG, Balan A, Souza Ferreira LCD. The Anti-Dengue Virus Peptide DV2 Inhibits Zika Virus Both In Vitro and In Vivo. Viruses 2023; 15:v15040839. [PMID: 37112820 PMCID: PMC10143277 DOI: 10.3390/v15040839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
The C-terminal portion of the E protein, known as stem, is conserved among flaviviruses and is an important target to peptide-based antiviral strategies. Since the dengue (DENV) and Zika (ZIKV) viruses share sequences in the stem region, in this study we evaluated the cross-inhibition of ZIKV by the stem-based DV2 peptide (419–447), which was previously described to inhibit all DENV serotypes. Thus, the anti-ZIKV effects induced by treatments with the DV2 peptide were tested in both in vitro and in vivo conditions. Molecular modeling approaches have demonstrated that the DV2 peptide interacts with amino acid residues exposed on the surface of pre- and postfusion forms of the ZIKA envelope (E) protein. The peptide did not have any significant cytotoxic effects on eukaryotic cells but efficiently inhibited ZIKV infectivity in cultivated Vero cells. In addition, the DV2 peptide reduced morbidity and mortality in mice subjected to lethal challenges with a ZIKV strain isolated in Brazil. Taken together, the present results support the therapeutic potential of the DV2 peptide against ZIKV infections and open perspectives for the development and clinical testing of anti-flavivirus treatments based on synthetic stem-based peptides.
Collapse
|
62
|
Leiva S, Bugnon Valdano M, Gardiol D. Unravelling the epidemiological diversity of Zika virus by analyzing key protein variations. Arch Virol 2023; 168:115. [PMID: 36943525 DOI: 10.1007/s00705-023-05726-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/19/2023] [Indexed: 03/23/2023]
Abstract
The consequences of Zika virus (ZIKV) infections were limited to sporadic mild diseases until almost a decade ago, when epidemic outbreaks took place, with quick spread into the Americas. Simultaneously, novel severe neurological manifestations of ZIKV infections were identified, including congenital microcephaly. However, why the epidemic strains behave differently is not yet completely understood, and many questions remain about the actual significance of genetic variations in the epidemiology and biology of ZIKV. In this study, we analysed a large number of viral sequences to identify genes with different levels of variability and patterns of genomic variations that could be associated with ZIKV diversity. We compared numerous epidemic strains with pre-epidemic strains, using the BWA-mem algorithm, and we also examined specific variations among the epidemic ZIKV strains derived from microcephaly cases. We identified several viral genes with dissimilar mutation rates among the ZIKV strain groups and novel protein variation profiles that might be associated with epidemiological particularities. Finally, we assessed the impact of the detected changes on the structure and stability of the NS1, NS5, and E proteins using the I-TASSER, trRosetta, and RaptorX modelling algorithms, and we found some interesting variations that might help to explain the heterogeneous features of the diverse ZIKA strains. This work contributes to the identification of genetic differences in the ZIKV genome that might have a phenotypic impact, providing a basis for future experimental analysis to elucidate the genetic causes of the recent ZIKV emergency.
Collapse
Affiliation(s)
- Santiago Leiva
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina
| | - Marina Bugnon Valdano
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| | - Daniela Gardiol
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Instituto de Biología Molecular y Celular de Rosario-CONICET, Universidad Nacional de Rosario, Suipacha 531, 2000, Rosario, Argentina.
| |
Collapse
|
63
|
Gaspar-Castillo C, Rodríguez MH, Ortiz-Navarrete V, Alpuche-Aranda CM, Martinez-Barnetche J. Structural and immunological basis of cross-reactivity between dengue and Zika infections: Implications in serosurveillance in endemic regions. Front Microbiol 2023; 14:1107496. [PMID: 37007463 PMCID: PMC10063793 DOI: 10.3389/fmicb.2023.1107496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/24/2023] [Indexed: 03/19/2023] Open
Abstract
Dengue and Zika are arthropod-borne viral diseases present in more than 100 countries around the world. In the past decade, Zika emerged causing widespread outbreaks in new regions, where dengue has been endemic-epidemic for a long period. The wide and extensive dissemination of the mosquito vectors, Aedes aegypti, and Ae. albopictus, favor the co-existence of both infections in the same regions. Together with an important proportion of asymptomatic infections, similar clinical manifestations, and a short time window for acute infection confirmatory tests, it is difficult to differentially estimate both dengue and Zika incidence and prevalence. DENV and ZIKV flavivirus share high structural similarity, inducing a cross-reactive immune response that leads to false positives in serological tests particularly in secondary infections. This results in overestimation of recent Zika outbreaks seroprevalence in dengue endemic regions. In this review, we address the biological basis underlying DENV and ZIKV structural homology; the structural and cellular basis of immunological cross reactivity; and the resulting difficulties in measuring dengue and Zika seroprevalence. Finally, we offer a perspective about the need for more research to improve serological tests performance.
Collapse
Affiliation(s)
- Carlos Gaspar-Castillo
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Mario H. Rodríguez
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City, Mexico
| | - Celia M. Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- Celia M. Alpuche-Aranda,
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, Cuernavaca, Mexico
- *Correspondence: Jesus Martinez-Barnetche,
| |
Collapse
|
64
|
Marano JM, Weger-Lucarelli J. Replication in the presence of dengue convalescent serum impacts Zika virus neutralization sensitivity and fitness. Front Cell Infect Microbiol 2023; 13:1130749. [PMID: 36968111 PMCID: PMC10034770 DOI: 10.3389/fcimb.2023.1130749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/13/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction Flaviviruses like dengue virus (DENV) and Zika virus (ZIKV) are mosquito-borne viruses that cause febrile, hemorrhagic, and neurological diseases in humans, resulting in 400 million infections annually. Due to their co-circulation in many parts of the world, flaviviruses must replicate in the presence of pre-existing adaptive immune responses targeted at serologically closely related pathogens, which can provide protection or enhance disease. However, the impact of pre-existing cross-reactive immunity as a driver of flavivirus evolution, and subsequently the implications on the emergence of immune escape variants, is poorly understood. Therefore, we investigated how replication in the presence of convalescent dengue serum drives ZIKV evolution. Methods We used an in vitro directed evolution system, passaging ZIKV in the presence of serum from humans previously infected with DENV (anti-DENV) or serum from DENV-naïve patients (control serum). Following five passages in the presence of serum, we performed next-generation sequencing to identify mutations that arose during passaging. We studied two non-synonymous mutations found in the anti-DENV passaged population (E-V355I and NS1-T139A) by generating individual ZIKV mutants and assessing fitness in mammalian cells and live mosquitoes, as well as their sensitivity to antibody neutralization. Results and discussion Both viruses had increased fitness in Vero cells with and without the addition of anti-DENV serum and in human lung epithelial and monocyte cells. In Aedes aegypti mosquitoes-using blood meals with and without anti-DENV serum-the mutant viruses had significantly reduced fitness compared to wild-type ZIKV. These results align with the trade-off hypothesis of constrained mosquito-borne virus evolution. Notably, only the NS1-T139A mutation escaped neutralization, while E-V335I demonstrated enhanced neutralization sensitivity to neutralization by anti-DENV serum, indicating that neutralization escape is not necessary for viruses passaged under cross-reactive immune pressures. Future studies are needed to assess cross-reactive immune selection in humans and relevant animal models or with different flaviviruses.
Collapse
Affiliation(s)
- Jeffrey M. Marano
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Roanoke, VA, United States
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
65
|
Prudencio CR, Gomes da Costa V, Rocha LB, da Costa HHM, Orts DJB, da Silva Santos FR, Rahal P, Lino NAB, da Conceição PJP, Bittar C, Machado RRG, Durigon EL, Araujo JP, Polatto JM, da Silva MA, de Oliveira JA, Mitsunari T, Pereira LR, Andreata-Santos R, de Souza Ferreira LC, Luz D, Piazza RMF. Identification of Zika Virus NS1-Derived Peptides with Potential Applications in Serological Tests. Viruses 2023; 15:v15030654. [PMID: 36992364 PMCID: PMC10052002 DOI: 10.3390/v15030654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/05/2023] Open
Abstract
Zika virus (ZIKV), a mosquito-borne pathogen, is an emerging arbovirus associated with sporadic symptomatic cases of great medical concern, particularly among pregnant women and newborns affected with neurological disorders. Serological diagnosis of ZIKV infection is still an unmet challenge due to the co-circulation of the dengue virus, which shares extensive sequence conservation of structural proteins leading to the generation of cross-reactive antibodies. In this study, we aimed to obtain tools for the development of improved serological tests for the detection of ZIKV infection. Polyclonal sera (pAb) and a monoclonal antibody (mAb 2F2) against a recombinant form of the ZIKV nonstructural protein 1 (NS1) allowed the identification of linear peptide epitopes of the NS1 protein. Based on these findings, six chemically synthesized peptides were tested both in dot blot and ELISA assays using convalescent sera collected from ZIKV-infected patients. Two of these peptides specifically detected the presence of ZIKV antibodies and proved to be candidates for the detection of ZIKV-infected subjects. The availability of these tools opens perspectives for the development of NS1-based serological tests with enhanced sensitivity regarding other flaviviruses.
Collapse
Affiliation(s)
- Carlos Roberto Prudencio
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Vivaldo Gomes da Costa
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Leticia Barboza Rocha
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Hernan Hermes Monteiro da Costa
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Diego José Belato Orts
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Felipe Rocha da Silva Santos
- Laboratório de Imunobiotecnologia, Centro de Imunologia, Instituto Adolfo Lutz, Av. Dr. Arnaldo, 351, São Paulo 01246-902, SP, Brazil
| | - Paula Rahal
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Nikolas Alexander Borsato Lino
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Pâmela Jóyce Previdelli da Conceição
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Cintia Bittar
- Instituto de Biociências Letras e Ciências Exatas, Universidade Estadual Paulista Júlio de Mesquita Filho, Rua Cristóvão Colombo, 2265, São Jose do Rio Preto 15054-000, SP, Brazil
| | - Rafael Rahal Guaragna Machado
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Edison Luiz Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - João Pessoa Araujo
- Instituto de Biotecnologia, Universidade Estadual Paulista Júlio de Mesquita Filho, Botucatu 18607-440, SP, Brazil
| | - Juliana Moutinho Polatto
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Miriam Aparecida da Silva
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Joyce Araújo de Oliveira
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Thais Mitsunari
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
| | - Lennon Ramos Pereira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Robert Andreata-Santos
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Luís Carlos de Souza Ferreira
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
- Plataforma Científica Pasteur USP, Universidade de São Paulo, São Paulo 05508-000, SP, Brazil
| | - Daniela Luz
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| | - Roxane Maria Fontes Piazza
- Laboratório de Bacteriologia, Instituto Butantan, Av. Vital Brazil, 1500, São Paulo 05503-900, SP, Brazil
- Correspondence: (C.R.P.); (D.L.); (R.M.F.P.); Tel.: +55-11-3068-2886 (C.R.P.); +55-11-2627-9708 (D.L.); +55-11-2627-9724 (R.M.F.P.)
| |
Collapse
|
66
|
Poveda Cuevas SA, Barroso da Silva FL, Etchebest C. NS1 from Two Zika Virus Strains Differently Interact with a Membrane: Insights to Understand Their Differential Virulence. J Chem Inf Model 2023; 63:1386-1400. [PMID: 36780300 DOI: 10.1021/acs.jcim.2c01461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Zika virus (ZIKV) from Uganda (UG) expresses a phenotype related to fetal loss, whereas the variant from Brazil (BR) induces microcephaly in neonates. The differential virulence has a direct relation to biomolecular mechanisms that make one strain more aggressive than the other. The nonstructural protein 1 (NS1) is a key viral toxin to comprehend these viral discrepancies because of its versatility in many processes of the virus life cycle. Here, we aim to examine through coarse-grained models and molecular dynamics simulations the protein-membrane interactions for both NS1ZIKV-UG and NS1ZIKV-BR dimers. A first evaluation allowed us to establish that the NS1 proteins, in the membrane presence, explore new conformational spaces when compared to systems simulated without a lipid bilayer. These events derive from both differential coupling patterns and discrepant binding affinities to the membrane. The N-terminal domain, intertwined loop, and greasy finger proposed previously as binding membrane regions were also computationally confirmed by us. The anchoring sites have aromatic and ionizable residues that manage the assembly of NS1 toward the membrane, especially for the Ugandan variant. Furthermore, in the presence of the membrane, the difference in the dynamic cross-correlation of residues between the two strains is particularly pronounced in the putative epitope regions. This suggests that the protein-membrane interaction induces changes in the distal part related to putative epitopes. Taken together, these results open up new strategies for the treatment of flaviviruses that would specifically target these dynamic differences.
Collapse
Affiliation(s)
- Sergio Alejandro Poveda Cuevas
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Goethe University Frankfurt, Institute of Biochemistry II, Theodor-Stern-Kai 7, Frankfurt am Main, Hesse DE-60590, Germany.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Fernando L Barroso da Silva
- Programa Interunidades em Bioinformática, Universidade de São Paulo, Rua do Matão, 1010, São Paulo, São Paulo BR-05508-090, Brazil.,Departamento de Ciências Biomoleculares, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av. do Café, s/no-Campus da USP, Ribeirão Preto, São Paulo BR-14040-903, Brazil.,Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| | - Catherine Etchebest
- Université Paris Cité and Université des Antilles, INSERM, Biologie Intégrée du Globule Rouge, F-75015 Paris, France.,Faculdade de Ciências Farmacêuticas de Ribeirão Preto, University of São Paulo and Université de Paris International Laboratory in Structural Bioinformatics, Av. do Café, s/no-Campus da USP, Bloco B, Ribeirão Preto, São Paulo BR-14040-903, Brazil
| |
Collapse
|
67
|
Benchimol GDC, Santos JB, Lopes ASDC, Oliveira KG, Okada EST, de Alcantara BN, Pereira WLA, Leão DL, Martins ACC, Carneiro LA, Imbeloni AA, Makiama ST, de Castro LPPA, Coutinho LN, Casseb LMN, Vasconcelos PFDC, Domingues SFS, Medeiros DBDA, Scalercio SRRDA. Zika Virus Infection Damages the Testes in Pubertal Common Squirrel Monkeys ( Saimiri collinsi). Viruses 2023; 15:615. [PMID: 36992324 PMCID: PMC10051343 DOI: 10.3390/v15030615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 02/26/2023] Open
Abstract
During the Zika virus (ZIKV) outbreak and after evidence of its sexual transmission was obtained, concerns arose about the impact of the adverse effects of ZIKV infection on human fertility. In this study, we evaluated the clinical-laboratory aspects and testicular histopathological patterns of pubertal squirrel monkeys (Saimiri collinsi) infected with ZIKV, analyzing the effects at different stages of infection. The susceptibility of S. collinsi to ZIKV infection was confirmed by laboratory tests, which detected viremia (mean 1.63 × 106 RNA copies/µL) and IgM antibody induction. Reduced fecal testosterone levels, severe testicular atrophy and prolonged orchitis were observed throughout the experiment by ultrasound. At 21 dpi, testicular damage associated with ZIKV was confirmed by histopathological and immunohistochemical (IHC) analyses. Tubular retraction, the degeneration and necrosis of somatic and germ cells in the seminiferous tubules, the proliferation of interstitial cells and an inflammatory infiltrate were observed. ZIKV antigen was identified in the same cells where tissue injuries were observed. In conclusion, squirrel monkeys were found to be susceptible to the Asian variant of ZIKV, and this model enabled the identification of multifocal lesions in the seminiferous tubules of the infected group evaluated. These findings may suggest an impact of ZIKV infection on male fertility.
Collapse
Affiliation(s)
- Gabriela da Costa Benchimol
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | - Josye Bianca Santos
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Postgraduate Program in Animal Reproduction in the Amazon (ReproAmazon), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
| | | | | | | | | | - Washington Luiz Assunção Pereira
- Laboratory of Animal Pathology (LABOPAT), Institute of Health and Animal Production, Federal Rural University of the Amazon, Belém 66077-830, Pará, Brazil
| | - Danuza Leite Leão
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Mamirauá Institute for Sustainable Development, Tefé 69553-225, Amazonas, Brazil
| | | | | | | | | | | | - Leandro Nassar Coutinho
- Laboratory of Animal Pathology (LABOPAT), Institute of Health and Animal Production, Federal Rural University of the Amazon, Belém 66077-830, Pará, Brazil
| | - Lívia Medeiros Neves Casseb
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Department of Pathology, Center of Biologic and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil
| | - Sheyla Farhayldes Souza Domingues
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Postgraduate Program in Animal Reproduction in the Amazon (ReproAmazon), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- School of Veterinary Medicine, Federal University of Pará, Castanhal 68740-970, Pará, Brazil
| | - Daniele Barbosa de Almeida Medeiros
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | | |
Collapse
|
68
|
Zephyr J, Rao DN, Johnson C, Shaqra AM, Nalivaika EA, Jordan A, Kurt Yilmaz N, Ali A, Schiffer CA. Allosteric quinoxaline-based inhibitors of the flavivirus NS2B/NS3 protease. Bioorg Chem 2023; 131:106269. [PMID: 36446201 PMCID: PMC10155214 DOI: 10.1016/j.bioorg.2022.106269] [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: 09/16/2022] [Revised: 10/27/2022] [Accepted: 11/06/2022] [Indexed: 11/20/2022]
Abstract
Viruses from the Flavivirus genus infect millions of people worldwide and cause severe diseases, including recent epidemics of dengue virus (DENV), and Zika virus (ZIKV). There is currently no antiviral treatment against flavivirus infections, despite considerable efforts to develop inhibitors against essential viral enzymes including NS2B/NS3 protease. Targeting the flavivirus NS2B/NS3 protease proved to be challenging because of the conformational dynamics, topology, and electrostatic properties of the active site. Here, we report the identification of quinoxaline-based allosteric inhibitors by fragment-based drug discovery approach as a promising new drug-like scaffold to target the NS2B/NS3 protease. Enzymatic assays and mutational analysis of the allosteric site in ZIKV NS2B/NS3 protease support noncompetitive inhibition mechanism as well as engineered DENV protease construct indicating the compounds likely compete with the NS2B cofactor for binding to the protease domain. Furthermore, antiviral activity confirmed the therapeutic potential of this new inhibitor scaffold.
Collapse
Affiliation(s)
- Jacqueto Zephyr
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Desaboini Nageswara Rao
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Colby Johnson
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Ala M Shaqra
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Ellen A Nalivaika
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Aria Jordan
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| | - Akbar Ali
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| |
Collapse
|
69
|
Gilbert RK, Petersen LR, Honein MA, Moore CA, Rasmussen SA. Zika virus as a cause of birth defects: Were the teratogenic effects of Zika virus missed for decades? Birth Defects Res 2023; 115:265-274. [PMID: 36513609 PMCID: PMC10552063 DOI: 10.1002/bdr2.2134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022]
Abstract
Zika virus (ZIKV) was identified as a teratogen in 2016 when an increase in severe microcephaly and other brain defects was observed in fetuses and newborns following outbreaks in French Polynesia (2013-2014) and Brazil (2015-2016) and among travelers to other countries experiencing outbreaks. Some have questioned why ZIKV was not recognized as a teratogen before these outbreaks: whether novel genetic changes in ZIKV had increased its teratogenicity or whether its association with birth defects had previously been undetected. Here we examine the evidence for these two possibilities. We describe evidence for specific mutations that arose before the French Polynesia outbreak that might have increased ZIKV teratogenicity. We also present information on children born with findings consistent with congenital Zika syndrome (CZS) as early as 2009 and epidemiological evidence that suggests increases in CZS-type birth defects before 2013. We also explore reasons why a link between ZIKV and birth defects might have been missed, including issues with surveillance of ZIKV infections and of birth defects, challenges to ZIKV diagnostic testing, and the susceptibility of different populations to ZIKV infection at the time of pregnancy. Although it is not possible to prove definitively that ZIKV had teratogenic properties before 2013, several pieces of evidence support the hypothesis that its teratogenicity had been missed in the past. These findings emphasize the need for further investments in global surveillance for emerging infections and for birth defects so that infectious teratogens can be identified more expeditiously in the future.
Collapse
Affiliation(s)
- Rachel K. Gilbert
- University of Florida College of Medicine, Gainesville, Florida, USA
| | - Lyle R. Petersen
- Division of Vector-Borne Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado, USA
| | - Margaret A. Honein
- Division of Preparedness and Emerging Infections, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Cynthia A. Moore
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Goldbelt Professional Services, LLC, Chesapeake, Virginia, USA
| | - Sonja A. Rasmussen
- Departments of Pediatrics and Obstetrics and Gynecology, College of Medicine, University of Florida, Gainesville, Florida, USA
- Department of Epidemiology, College of Medicine and College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
70
|
Warner JC, Hatziioanou D, Osborne JC, Bailey DJ, Brooks TJG, Semper AE. Infections in travellers returning to the UK: a retrospective analysis (2015-2020). J Travel Med 2023; 30:7008452. [PMID: 36708032 DOI: 10.1093/jtm/taad003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Every year, many thousands of travellers return to the United Kingdom (UK) from visits to other countries and some will become unwell due to infections acquired abroad. Many imported infections have similar clinical presentations, such as fever and myalgia, so diagnostic testing is an important tool to improve patient management and outcomes. The aim of this study was to examine the demographics, travel history, presenting symptoms and diagnostic outcomes of referrals to the UK's specialist diagnostic Rare & Imported Pathogens Laboratory (RIPL) for the period 2015-2020. METHODS Anonymised clinical and laboratory data was extracted from RIPL's Laboratory Information Management System (LIMS) and cleaned prior to descriptive analysis of the data. Travel history data was mapped to one of eight world regions, while symptom data was categorised into presenting syndromes. Diagnostic data was categorised as either positive, equivocal or negative. RESULTS During the period 2015-2020, RIPL received 73 951 samples from 53 432 patients suspected of having infections that are rare in the UK. The most common age group for unwell returning travellers was 30-39 years and the most commonly reported travel destination was Southern and SE Asia. Dengue virus was the most diagnosed infection overall, followed by chikungunya, Zika, leptospirosis and spotted fever group Rickettsia. Dengue virus was among the top three most frequent diagnoses for all world regions except Europe and represented 62.5% of all confirmed/probable diagnoses. CONCLUSIONS None of the top five infections diagnosed by RIPL in travellers are vaccine-preventable, therefore understanding traveller demographics, destination-specific risk factors and encouraging preventative behaviours is the best available strategy to reduce the number of returning travellers who become infected. Prompt referral of acute samples with a detailed travel history, including purpose of travel and activities undertaken as well as dates and destinations can be a valuable tool in designing public health interventions and diagnostic algorithms.
Collapse
Affiliation(s)
- Jennifer C Warner
- Rare & Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, UK
| | | | - Jane C Osborne
- Rare & Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, UK
| | - Daniel J Bailey
- Diagnostic Support, UK Health Security Agency, Porton Down, UK
| | - Timothy J G Brooks
- Rare & Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, UK
| | - Amanda E Semper
- Rare & Imported Pathogens Laboratory, UK Health Security Agency, Porton Down, UK
| |
Collapse
|
71
|
Delfin-Riela T, Rossotti MA, Mattiuzzo G, Echaides C, González-Sapienza G. Nanobody-Based Blocking of Binding ELISA for the Detection of Anti-NS1 Zika-Virus-Specific Antibodies in Convalescent Patients. Trop Med Infect Dis 2023; 8:tropicalmed8010055. [PMID: 36668962 PMCID: PMC9862682 DOI: 10.3390/tropicalmed8010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/13/2023] Open
Abstract
Zika virus has spread around the world with rapid pace in the last five years. Although symptoms are typically mild and unspecific, Zika's major impact occurs during pregnancy, generating a congenital syndrome. Serology plays a key role in its diagnosis. However, its use is limited due to the uncertainty caused by the cross-reaction of antibodies elicited in response to other flavivirus infections when tested in direct immunoassays. Using a panel of previously generated anti-Zika non-structural protein 1 (NS1) nanobodies, a set was selected that only recognizes epitopes present in Zika and is immunogenic to humans. A proper arrangement of these nanobodies was made and conditions were optimized in order to develop a novel serology assay. This new ELISA relies on the inhibition of the binding of a set of selected nanobodies to Zika-immobilized NS1 when previously incubated with Zika convalescent sera. Using the developed blocking of binding assay, it was possible to discriminate between Zika-specific and cross-reactive antibodies in serum samples from infections with Zika and other flaviviruses.
Collapse
Affiliation(s)
- Triana Delfin-Riela
- Cátedra de Inmunología, DEPBIO, Facultad de Química, Instituto de Higiene, UDELAR, Montevideo 11600, Uruguay
| | - Martín A. Rossotti
- Cátedra de Inmunología, DEPBIO, Facultad de Química, Instituto de Higiene, UDELAR, Montevideo 11600, Uruguay
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control (NIBSC)-MHRA, Hertfordshire EN6 3QG, UK
| | | | - Gualberto González-Sapienza
- Cátedra de Inmunología, DEPBIO, Facultad de Química, Instituto de Higiene, UDELAR, Montevideo 11600, Uruguay
- Correspondence: ; Tel.: +598-24874334
| |
Collapse
|
72
|
Touchton M, Wampler B. Democratizing Public Health: Participatory Policymaking Institutions, Mosquito Control, and Zika in the Americas. Trop Med Infect Dis 2023; 8:tropicalmed8010038. [PMID: 36668945 PMCID: PMC9865320 DOI: 10.3390/tropicalmed8010038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
The Zika virus is a mosquito-borne virus spread primarily by Aedes mosquitoes. Zika cases have been detected throughout the mosquito's range, with an epidemic occurring from 2015 to 2017 in Brazil. Many Zika cases are mild or asymptomatic, but infections in pregnant women can cause microcephaly in children, and a small percentage of cases result in Guillan-Barré syndrome. There is currently little systematic information surrounding the municipal spread of the Zika Virus in Brazil. This article uses coarsened exact matching with negative binomial estimation and ordinary least squares estimation to assess the determinants of Zika incidence across the ~280,000 cases confirmed and recorded by Brazil's Ministry of Health in 2016 and 2017. These data come from Freedom of Information Act (FOIA) requests in Brazil and have not been published. We use data on the universe of individual Zika cases in Brazil and Geographic Information Systems (GIS) software to examine the virus at the municipal level across 5570 municipalities and construct a unique, unusually rich dataset covering daily Zika transmission. Additionally, our dataset includes corresponding local data on democratic governance, mosquito control efforts, and environmental conditions to estimate their relationship to Zika transmission. The results demonstrate that the presence of subnational democratic, participatory policymaking institutions and high levels of local state capacity are associated with low rates of Zika contraction. These models control for local healthcare spending and economic conditions, among other factors, that also influence Zika contraction rates. In turn, these findings provide a better understanding of what works for local health governance and mosquito control and makes important data public so that scholars and practitioners can perform their own analyses. Stronger models of Zika transmission will then inform mosquito abatement efforts across the Global South, as well as provide a blueprint for combatting Dengue fever, which is also transmitted by Aedes mosquitoes.
Collapse
Affiliation(s)
- Michael Touchton
- Department of Political Science, University of Miami, Coral Gables, FL 33146, USA
- Faculty Lead for Global Health, Institute for Advanced Studies of the Americas, University of Miami, Coral Gables, FL 33146, USA
- Correspondence:
| | - Brian Wampler
- President’s Office of Public Engagement, Boise State University, Boise, ID 83725, USA
| |
Collapse
|
73
|
Hu D, Zou H, Chen W, Li Y, Luo Z, Wang X, Guo D, Meng Y, Liao F, Wang W, Zhu Y, Wu J, Li G. ZDHHC11 Suppresses Zika Virus Infections by Palmitoylating the Envelope Protein. Viruses 2023; 15:144. [PMID: 36680184 PMCID: PMC9863066 DOI: 10.3390/v15010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Zika virus (ZIKV) is an RNA-enveloped virus that belongs to the Flavivirus genus, and ZIKV infections potentially induce severe neurodegenerative diseases and impair male fertility. Palmitoylation is an important post-translational modification of proteins that is mediated by a series of DHHC-palmitoyl transferases, which are implicated in various biological processes and viral infections. However, it remains to be investigated whether palmitoylation regulates ZIKV infections. In this study, we initially observed that the inhibition of palmitoylation by 2-bromopalmitate (2-BP) enhanced ZIKV infections, and determined that the envelope protein of ZIKV is palmitoylated at Cys308. ZDHHC11 was identified as the predominant enzyme that interacts with the ZIKV envelope protein and catalyzes its palmitoylation. Notably, ZDHHC11 suppressed ZIKV infections in an enzymatic activity-dependent manner and ZDHHC11 knockdown promoted ZIKV infection. In conclusion, we proposed that the envelope protein of ZIKV undergoes a novel post-translational modification and identified a distinct mechanism in which ZDHHC11 suppresses ZIKV infections via palmitoylation of the ZIKV envelope protein.
Collapse
Affiliation(s)
- Dingwen Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Haimei Zou
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Weijie Chen
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Yuting Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziqing Luo
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xianyang Wang
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Dekuan Guo
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yu Meng
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Feng Liao
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wenbiao Wang
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ying Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Geng Li
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
74
|
The size of larval rearing container modulates the effects of diet amount and larval density on larval development in Aedes aegypti. PLoS One 2023; 18:e0280736. [PMID: 36696416 PMCID: PMC9876358 DOI: 10.1371/journal.pone.0280736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 01/06/2023] [Indexed: 01/26/2023] Open
Abstract
Mass-rearing of mosquitoes under laboratory conditions is an important part of several new control techniques that rely on the release of males to control mosquito populations. While previous work has investigated the effect of larval density and diet amount on colony productivity, the role of the size of the container in which larval development takes place has been relatively ignored. We investigated the role of container size in shaping life history and how this varied with density and food availability in Aedes aegypti, an important disease vector and target of mass-rearing operations. For each treatment combination, immature development time and survival and adult body size and fecundity were measured, and then combined into a measure of productivity. We additionally investigated how larval aggregation behaviour varied with container size. Container size had important effects on life history traits and overall productivity. In particular, increasing container size intensified density and diet effects on immature development time. Productivity was also impacted by container size when larvae were reared at high densities (1.4 larva/ml). In these treatments, the productivity metric of large containers was estimated to be significantly lower than medium or small containers. Regardless of container size, larvae were more likely to be observed at the outer edges of containers, even when this led to extremely high localized densities. We discuss how container size and larval aggregation responses may alter the balance of energy input and output to shape development and productivity.
Collapse
|
75
|
Yu Y, Chen Y, Wang J, Fan X, He Z, Qiao S, Hou S, Zou P. A peptide derived from the N-terminal of NS2A for the preparation of ZIKV NS2A recognition polyclonal antibody. J Immunol Methods 2023; 512:113396. [PMID: 36463933 DOI: 10.1016/j.jim.2022.113396] [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: 06/14/2022] [Revised: 11/10/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Zika virus non-structural protein NS2A participates in viral replication, organization, and budding, as well as escaping host immunity. NS2A also involved in the induction of microcephaly by ZIKV. However, the above studies were mainly performed through NS2A with a tag due to the lack of available antibodies against NS2A. ZIKV NS2A is a multiplex transmembrane protein, which leads to difficulties in the preparation of its recognition antibodies, thus seriously affecting the study of ZIKV NS2A. In this study, we found that a peptide (GSTDHMDHFSLGVLC) derived from the N-terminal of ZIKV NS2A coupled to KLH induced antibodies recognizing ZIKV NS2A in rabbits. The purified polyclonal antibody recognized ZIKV NS2A in ZIKV-infected cells with high efficiency and specificity, as detected by western blot and immunofluorescence assay. Our study has important implications for the preparation of ZIKV NS2A antibodies and the in-depth study of ZIKV NS2A.
Collapse
Affiliation(s)
- Yufeng Yu
- Shanxi Provincial Key Laboratory for Functional Proteins, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China.
| | - Yongkang Chen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Jian Wang
- Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd., Taiyuan 030032, China
| | - Xiuling Fan
- Shanxi Provincial Key Laboratory for Functional Proteins, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, China
| | - Zhenrui He
- Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd., Taiyuan 030032, China
| | - Shaojun Qiao
- Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd., Taiyuan 030032, China
| | - Shishi Hou
- Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd., Taiyuan 030032, China
| | - Peng Zou
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China.
| |
Collapse
|
76
|
Costa ACD, Morais VDS, Azevedo RMD, Nuevo KMB, Cunha MS. Genetic characterization of the rare Bruconha virus (Bunyavirales: Orthobunyavirus) isolated in Vale do Ribeira (Atlantic Forest biome), Southeastern Brazil. Rev Inst Med Trop Sao Paulo 2023; 65:e17. [PMID: 36921205 PMCID: PMC10013467 DOI: 10.1590/s1678-9946202365017] [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: 09/16/2022] [Accepted: 01/24/2023] [Indexed: 03/17/2023] Open
Abstract
Brazil is a great source of arbovirus diversity, mainly in the Amazon region. However, other biomes, especially the Atlantic Forest, may also be a hotspot for emerging viruses, including Bunyaviruses (Negarnaviricota: Bunyavirales). For instance, Vale do Ribeira, located in the Southeastern region, has been widely studied for virus surveillance, where Flavivirus, Alphavirus and Bunyaviruses were isolated during the last decades, including Bruconha virus (BRCV), a member of Orthobunyavirus genus Group C, in 1976. Recently, a new isolate of BRCV named Span321532 was obtained from an adult sentinel mouse placed in Iguape city in 2011, and a full-length genome was generated with nucleotide differences ranging between 1.5%, 5.3% and 5% (L, M and S segments, respectively) from the prototype isolated 35 years earlier. In addition, each segment placed BRCV into different clusters, showing the high variety within Bunyavirales. Although no evidence for reassortants was detected, this finding reiterates the need for new surveillance and genomic studies in the area considering the high mutation rates of arbovirus, and also to identify the hosts capable of supporting the continuous circulation of Orthobunyavirus.
Collapse
Affiliation(s)
- Antônio Charlys da Costa
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | - Vanessa Dos Santos Morais
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | - Roberta Marcatti de Azevedo
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Mariana Sequetin Cunha
- Universidade de São Paulo, Faculdade de Medicina, Instituto de Medicina Tropical de São Paulo, São Paulo, São Paulo, Brazil.,Instituto Adolfo Lutz, Núcleo de Doenças de Transmissão Vetorial, São Paulo, São Paulo, Brazil
| |
Collapse
|
77
|
SARS-CoV-2 Genomic Surveillance in Brazil: A Systematic Review with Scientometric Analysis. Viruses 2022; 14:v14122715. [PMID: 36560720 PMCID: PMC9784312 DOI: 10.3390/v14122715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022] Open
Abstract
Several studies have monitored the SARS-CoV-2 variants in Brazil throughout the pandemic. Here, we systematically reviewed and conducted a scientometric analysis of the SARS-CoV-2 genomic surveillance studies using Brazilian samples. A Pubmed database search on October 2022 returned 492 articles, of which 106 were included. Ninety-six different strains were reported, with variant of concern (VOC) gamma (n = 35,398), VOC delta (n = 15,780), and the variant of interest zeta (n = 1983) being the most common. The top three states with the most samples in the published articles were São Paulo, Rio de Janeiro, and Minas Gerais. Whereas the first year of the pandemic presented primary circulation of B.1.1.28 and B.1.1.33 variants, consecutive replacements were observed between them and VOI zeta, VOC gamma, VOC delta, and VOC omicron. VOI mu, VOI lambda, VOC alpha, and VOC beta were also detected but failed to reach significant circulation. Co-infection, re-infection, and vaccine breakthrough reports were found. Article co-citation differed from the co-authorship structure. Despite the limitations, we expect to give an overview of Brazil's genomic surveillance studies and contribute to future research execution.
Collapse
|
78
|
Stein RA, Grayon A, Katz A, Chervenak FA. The Zika virus: an opportunity to revisit reproductive health needs and disparities. Germs 2022; 12:519-537. [PMID: 38021183 PMCID: PMC10660223 DOI: 10.18683/germs.2022.1357] [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: 08/30/2022] [Revised: 10/24/2022] [Accepted: 12/29/2022] [Indexed: 12/01/2023]
Abstract
First isolated in 1947, the Zika virus was initially connected only to limited or sporadic human infections. In late 2015, the temporal clustering of a Zika outbreak and microcephaly in newborn babies from northeastern Brazil, and the identification of a causal link between the two, led to the characterization of the congenital Zika syndrome. In the wake of the epidemic, several countries from Latin America advised women to postpone pregnancies for periods ranging from six months to two years. These recommendations initiated critical conversations about the challenges of implementing them in societies with limited access to contraception, widespread socioeconomic inequalities, and high rates of unplanned and adolescent pregnancies. The messaging targeted exclusively women, despite a high prevalence of imbalances in the relationship power, and addressed all women as a group, failing to recognize that the decision to postpone pregnancies will impact different women in different ways, depending on their age at the time. Finally, in several countries affected by the Zika epidemic, due to restrictive reproductive policies, legally terminating a pregnancy is no longer an option even at the earliest time when brain malformations as part of the congenital Zika syndrome can be detected by ultrasonography. The virus continued to circulate after 2016 in several countries. Climate change models predict an expansion of the geographical area where local Zika transmission may occur, indicating that the interface between the virus, teratogenesis, and reproductive rights is a topic of considerable interest for medicine, social sciences, and public health for years to come.
Collapse
Affiliation(s)
- Richard A. Stein
- MD, PhD, NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn 11201, NY, USA
| | - Alexis Grayon
- NYU Tandon School of Engineering, Department of Chemical and Biomolecular Engineering, 6 MetroTech Center, Brooklyn 11201, NY, USA
| | - Adi Katz
- MD, Department of Obstetrics and Gynecology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 110 E 77th Street, New York, NY, 10075, USA
| | - Frank A. Chervenak
- MD, Department of Obstetrics and Gynecology, Lenox Hill Hospital, Donald and Barbara Zucker School of Medicine at Hofstra Northwell, 110 E 77th Street, New York, NY, 10075, USA
| |
Collapse
|
79
|
Evans TS, Aung O, Cords O, Coffey LL, Wong T, Weiss CM, Maw MT, Yee J, Venkateswaran K, Venkateswaran N, Nham P, Van Rompay KKA, Morris MK, Oceguera L, Werthimer W, Hanson C, Valitutto M, Tun KYN, Win YT, Thein WZ, Murray S, Thu HM, Johnson CK. Sylvatic Transmission of Chikungunya Virus among Nonhuman Primates in Myanmar. Emerg Infect Dis 2022; 28:2548-2551. [PMID: 36417997 PMCID: PMC9707571 DOI: 10.3201/eid2812.220893] [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] [Indexed: 12/13/2022] Open
Abstract
Nonhuman primates living in proximity to humans increase risks for sylvatic arbovirus transmission. We collected serum samples from nonhuman primates in Hlawga National Park near Yangon, Myanmar, and detected antibodies against chikungunya (33%) and Japanese encephalitis (4%) viruses. Buffer zones between primate and human communities might reduce cross-species arbovirus transmission.
Collapse
|
80
|
Liu J, Mao Y, Li Q, Qiu Z, Li J, Li X, Liang W, Xu M, Li A, Cai X, Wu W, Chen H, Yan R, Li J, Gu W, Li H. Efficient Gene Transfer to Kidney Using a Lentiviral Vector Pseudotyped with Zika Virus Envelope Glycoprotein. Hum Gene Ther 2022; 33:1269-1278. [PMID: 35904396 DOI: 10.1089/hum.2022.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gene therapy's entrance into clinical settings has made it an ever more attractive field of study for various diseases. However, relatively little progress has been made in targeting kidney diseases due to poor gene delivery efficiency in renal cells. The development of novel gene therapy vectors for medical intervention to treat kidney diseases is needed. In this study, we designed and produced a pseudotyped lentiviral vector with envelope glycoproteins of Zika virus (ZIKV), and evaluated its potential use in viral vector entry, neutralization assay, and gene delivery especially in the renal context. The lentiviral vector, simplified as ZIKV-E, is pseudotyped with Env/G-TC representing the transmembrane (TM) and cytoplasmic (CY) domains of Env replaced with the TM and CY domains of the glycoprotein (G) of the vesicular stomatitis virus. In vivo results show that ZIKV-E induced efficient transduction in tubular epithelial cells in mouse kidneys, demonstrating >100-fold higher expression of exogenous green fluorescent protein gene compared with that achieved by vesicular stomatitis virus G (VSV-G) protein pseudotyped lentiviral vector. The results also showed that the vector ZIKV-E transduced cells in a pH-independent manner and the transduction was inhibited by anti-ZIKV Env domain III antibodies. Results also show that ZIKV-E can be used as a surrogate for studies of ZIKV entry mechanisms and neutralization antibody assay. In all, this study successfully demonstrated a novel pseudotyped lentiviral vector ZIKV-E for inducing high transduction efficiency in renal tubular epithelial cells that could serve as a foundation for gene therapy for the treatment of inherited renal diseases in humans.
Collapse
Affiliation(s)
- Jun Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Institute of Dermatology and Venereology, Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yingying Mao
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Qingqing Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Clinical Laboratory, Foshan Women and Children Hospital, Foshan, China
| | - Zhenzhen Qiu
- Guangzhou Bioneeds Biotechnology Co., Ltd, Guangzhou, China
| | - Jingjing Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoxin Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wenhan Liang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Mingyu Xu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Andrew Li
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiangsheng Cai
- Center for Medical Experiments, University of Chinese Academy of Science-Shenzhen Hospital, Shenzhen, China; and
| | - Wangsheng Wu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Huangyao Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Renhe Yan
- Guangzhou Bioneeds Biotechnology Co., Ltd, Guangzhou, China
| | - Jinlong Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Weiwang Gu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Hongwei Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
81
|
Ling-Hu T, Rios-Guzman E, Lorenzo-Redondo R, Ozer EA, Hultquist JF. Challenges and Opportunities for Global Genomic Surveillance Strategies in the COVID-19 Era. Viruses 2022; 14:2532. [PMID: 36423141 PMCID: PMC9698389 DOI: 10.3390/v14112532] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/19/2022] Open
Abstract
Global SARS-CoV-2 genomic surveillance efforts have provided critical data on the ongoing evolution of the virus to inform best practices in clinical care and public health throughout the pandemic. Impactful genomic surveillance strategies generally follow a multi-disciplinary pipeline involving clinical sample collection, viral genotyping, metadata linkage, data reporting, and public health responses. Unfortunately, current limitations in each of these steps have compromised the overall effectiveness of these strategies. Biases from convenience-based sampling methods can obfuscate the true distribution of circulating variants. The lack of standardization in genotyping strategies and bioinformatic expertise can create bottlenecks in data processing and complicate interpretation. Limitations and inconsistencies in clinical and demographic data collection and sharing can slow the compilation and limit the utility of comprehensive datasets. This likewise can complicate data reporting, restricting the availability of timely data. Finally, gaps and delays in the implementation of genomic surveillance data in the public health sphere can prevent officials from formulating effective mitigation strategies to prevent outbreaks. In this review, we outline current SARS-CoV-2 global genomic surveillance methods and assess roadblocks at each step of the pipeline to identify potential solutions. Evaluating the current obstacles that impede effective surveillance can improve both global coordination efforts and pandemic preparedness for future outbreaks.
Collapse
Affiliation(s)
- Ted Ling-Hu
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL 60611, USA
| | - Estefany Rios-Guzman
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL 60611, USA
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL 60611, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL 60611, USA
| | - Judd F. Hultquist
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Center for Pathogen Genomics and Microbial Evolution, Robert J. Havey, MD Institute for Global Health, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
82
|
Lamb MM, Paniagua-Avila A, Zacarias A, Rojop N, Chacon A, Natrajan MS, Waggoner JJ, Lopez MR, Cordon-Rosales C, Huleatt JW, Bonaparte MI, Asturias EJ, Olson D. Repeated Rapid Active Sampling Surveys Demonstrated a Rapidly Changing Zika Seroprevalence among Children in a Rural Dengue-endemic Region in Southwest Guatemala during the Zika Epidemic (2015-2016). Am J Trop Med Hyg 2022; 107:1099-1106. [PMID: 36252798 PMCID: PMC9709015 DOI: 10.4269/ajtmh.22-0399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/08/2022] [Indexed: 11/07/2022] Open
Abstract
Although Central America is largely dengue virus (DENV)-endemic, the 2015-2016 Zika virus (ZIKV) pandemic brought new urgency to develop surveillance approaches capable of characterizing the rapidly changing disease burden in resource-limited settings. We conducted a pediatric DENV surveillance study in rural Guatemala, including serial cross-sectional surveys from April through September 2015 (Survey 1), in October-November 2015 (Survey 2), and January-February 2016 (Survey 3). Serum underwent DENV IgM MAC ELISA and polymerase chain reaction testing. Using banked specimens from Surveys 2 and 3, we expanded testing to include DENV 1-4 and ZIKV microneutralization (MN50), DENV NS1 IgG ELISA, and ZIKV anti-NS1 antibody Blockage of Binding (BoB) ELISA testing. Demographic risk factors for ZIKV BoB positivity were explored using multivariable generalized linear regression models. Of Survey 2 and 3 samples available (N = 382), DENV seroprevalence slightly increased (+1%-10% depending on the assay) during the surveillance period and increased with age. In contrast, ZIKV seroprevalence consistently increased over the 3-month period, including from 6% to 34% (P < 0.0001) and 10%-37% (P < 0.0001) using the MN50 ≥100 and BoB ELISA assays, respectively. Independent risk factors for ZIKV seropositivity included older age (prevalence ratio (PR)/year = 1.12, 95% confidence interval (CI) = 1.07-1.17) and primary caregiver literacy (PR = 2.80, CI = 1.30-6.06). Rapid active surveillance (RAS) surveys demonstrated a nearly 30% increase in ZIKV prevalence and a slight (≤ 10%) increase in DENV seroprevalence from October to November 2015 to January to February 2016 in rural southwest Guatemala, regardless of serologic assay used. RAS surveys may be a useful "off-the-shelf" tool to characterize arboviruses and other emerging pathogens rapidly in resource-limited settings.
Collapse
Affiliation(s)
- Molly M. Lamb
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, Aurora, Colorado
| | - Alejandra Paniagua-Avila
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
- Center for Human Development, Fundacion para la Salud Integral de los Guatemaltecos, FUNSALUD, Coatepeque, Quetzaltenango, Guatemala
| | - Alma Zacarias
- Center for Human Development, Fundacion para la Salud Integral de los Guatemaltecos, FUNSALUD, Coatepeque, Quetzaltenango, Guatemala
| | - Neudy Rojop
- Center for Human Development, Fundacion para la Salud Integral de los Guatemaltecos, FUNSALUD, Coatepeque, Quetzaltenango, Guatemala
| | - Andrea Chacon
- Center for Human Development, Fundacion para la Salud Integral de los Guatemaltecos, FUNSALUD, Coatepeque, Quetzaltenango, Guatemala
| | - Muktha S. Natrajan
- Emory University Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
| | - Jesse J. Waggoner
- Emory University Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia
| | - Maria Renee Lopez
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | - Celia Cordon-Rosales
- Centro de Estudios en Salud, Universidad del Valle de Guatemala, Guatemala City, Guatemala
| | | | | | - Edwin J. Asturias
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, Aurora, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Daniel Olson
- Department of Epidemiology and Center for Global Health, Colorado School of Public Health, Aurora, Colorado
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
83
|
Wang Y, Ling L, Zhang Z, Marin-Lopez A. Current Advances in Zika Vaccine Development. Vaccines (Basel) 2022; 10:vaccines10111816. [PMID: 36366325 PMCID: PMC9694033 DOI: 10.3390/vaccines10111816] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Zika virus (ZIKV), an emerging arthropod-borne flavivirus, was first isolated in Uganda in 1947 from monkeys and first detected in humans in Nigeria in 1952; it has been associated with a dramatic burden worldwide. Since then, interventions to reduce the burden of ZIKV infection have been mainly restricted to mosquito control, which in the end proved to be insufficient by itself. Hence, the situation prompted scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elucidated. Understanding the viral disease mechanism will provide a better landscape to develop prophylactic and therapeutic strategies against ZIKV. Currently, no specific vaccines or drugs have been approved for ZIKV. However, some are undergoing clinical trials. Notably, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, viral vectors, virus-like particles (VLPs), inactivated virus, live attenuated virus, peptide and protein-based vaccines, passive immunizations by using monoclonal antibodies (MAbs), and vaccines that target vector-derived antigens. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA titers, both in vitro and in vivo. This review provides a comprehensive summary of current advancements in the development of vaccines against Zika virus.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
- Correspondence:
| | - Lin Ling
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Zilei Zhang
- Department of Inspection and Quarantine Technology Communication, Shanghai Customs College, Shanghai 201204, China
| | - Alejandro Marin-Lopez
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06420, USA
| |
Collapse
|
84
|
Guo Y, Guo J, Li Y. Wolbachia wPip Blocks Zika Virus Transovarial Transmission in Aedes albopictus. Microbiol Spectr 2022; 10:e0263321. [PMID: 35894613 PMCID: PMC9603370 DOI: 10.1128/spectrum.02633-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/14/2022] [Indexed: 01/04/2023] Open
Abstract
Wolbachia is being developed as a biological tool to suppress mosquito populations and/or interfere with their transmitted viruses. Adult males with an artificial Wolbachia infection have been released, successfully yielding population suppression in multiple field trials. The main characteristic of the artificial Wolbachia-infected mosquitoes used in the suppression program is the lower vector competence than that in native infected/uninfected mosquitoes in horizontal and vertical transmission. Our previous studies have demonstrated that the Aedes albopictus HC line infected with a trio of Wolbachia strains exhibited almost complete blockade of dengue virus (DENV) and Zika virus (ZIKV) in horizontal and vertical transmission. However, the extent to which Wolbachia inhibits virus transovarial transmission is unknown since no studies have been performed to determine whether Wolbachia protects ovarian cells against viral infection. Here, we employed ovarian cells of the Ae. albopictus GUA (a wild-type mosquito line superinfected with two native Wolbachia strains, wAlbA and wAlbB), HC, and GT lines (tetracycline-cured, Wolbachia-uninfected mosquitoes), which exhibit key traits, and compared them to better understand how Wolbachia inhibits ZIKV transovarial transmission. Our results showed that the infection rate of adult GT progeny was significantly higher than that of GUA progeny during the first and second gonotrophic cycles. In contrast, the infection rates of adult GT and GUA progeny were not significantly different during the third gonotrophic cycle. All examined adult HC progeny from three gonotrophic cycles were negative for ZIKV infection. A strong negative linear correlation existed between Wolbachia density and ZIKV load in the ovaries of mosquitoes. Although there is no obvious coexistence area in the ovaries for Wolbachia and ZIKV, host immune responses may play a role in Wolbachia blocking ZIKV expansion and maintenance in the ovaries of Ae. albopictus. These results will aid in understanding Wolbachia-ZIKV interactions in mosquitoes. IMPORTANCE Area-wide application of Wolbachia to suppress mosquito populations and their transmitted viruses has achieved success in multiple countries. However, the mass release of Wolbachia-infected male mosquitoes involves a potential risk of accidentally releasing fertile females. In this study, we employed ovarian cells of the Ae. albopictus GUA, HC, and GT lines, which exhibit key traits, and compared them to better understand how Wolbachia inhibits ZIKV transovarial transmission. Our results showed an almost complete blockade of ZIKV transmission in HC female mosquitoes. Wolbachia in natively infected GUA mosquitoes negative affected ZIKV, and this interference was shown by slightly lower loads than those in HC mosquitoes. Overall, our work helps show how Wolbachia blocks ZIKV expansion and maintenance in the ovaries of Ae. albopictus and aids in understanding Wolbachia-ZIKV interactions in mosquitoes.
Collapse
Affiliation(s)
- Yan Guo
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Science, Guangzhou, Guangdong, China
| | - Jiatian Guo
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yifeng Li
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Science, Guangzhou, Guangdong, China
| |
Collapse
|
85
|
Wu Y, Yang R, Wu Q, Huang M, Shu B, Wu W, Sun B, Xia J, Chen X, Liao Y. Trace Analysis of Emerging Virus: An Ultrasensitive ECL-Scan Imaging System for Viral Infectious Disease. ACS OMEGA 2022; 7:37499-37508. [PMID: 36312431 PMCID: PMC9609065 DOI: 10.1021/acsomega.2c04280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/04/2022] [Indexed: 06/16/2023]
Abstract
Emerging infectious diseases have brought a huge impact on human society in recent years. The outbreak of Zika virus (ZIKV) in the Americas resulted in a large number of babies born with microcephaly. More seriously, the Coronavirus Disease 2019 (COVID-19) was globally spread and caused immeasurable damages. Thus, the monitoring of highly pathogenic viruses is important to prevent and control emerging infectious diseases. Herein, a dendritic polymer probe-amplified ECL-scan imaging system was constructed to realize trace analysis of viral emerging infectious diseases. A dendritic polymer probe was employed as the efficient signal emitter component that could generate an amplified ECL signal on the integrated chip, and the signal was detected by a single-photon level charge coupled device-based ECL-scan imaging system. With this strategy, the ZIKV in a complex system of blood, urine, and saliva was detected. The results indicated that a high sensitivity of 50 copies and superior specificity were achieved. Furthermore, this strategy realized highly sensitive detection (10 copies) of the S and N protein gene sequence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-Cov2) and spiked pseudovirus samples. Thus, the dendritic polymer probe-amplified ECL-scan imaging system suitably met the strict clinical requirements for trace analysis of an emerging virus, and thus has the potential to serve as a paradigm for monitoring emerging infectious diseases.
Collapse
Affiliation(s)
- Yunxia Wu
- Department
of Burn Surgery & Department of Clinical Laboratory, First People’s Hospital of Foshan, Foshan 528000, China
| | - Ronghua Yang
- Department
of Burn and Plastic Surgery, Guangzhou First
People’s Hospital, Guangzhou 510180, China
| | - Qikang Wu
- Department
of Burn Surgery & Department of Clinical Laboratory, First People’s Hospital of Foshan, Foshan 528000, China
| | - Mingxing Huang
- Department
of Infectious Disease, Fifth Affiliated
Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Bowen Shu
- Molecular
Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, China
| | - Wenjie Wu
- Molecular
Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, China
| | - Baoqing Sun
- Guangzhou
Institute of Respiratory Health, State Key Laboratory of Respiratory
Disease, National Clinical Research Center of Respiratory Disease, First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Jinyu Xia
- Department
of Infectious Disease, Fifth Affiliated
Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Xiaodong Chen
- Department
of Burn Surgery & Department of Clinical Laboratory, First People’s Hospital of Foshan, Foshan 528000, China
| | - Yuhui Liao
- Molecular
Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou 510091, China
- Department
of Infectious Disease, Fifth Affiliated
Hospital of Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
86
|
Lebeau G, El Safadi D, Paulo-Ramos A, Hoareau M, Desprès P, Krejbich-Trotot P, Chouchou F, Roche M, Viranaicken W. The Efficient Antiviral Response of A549 Cells Is Enhanced When Mitochondrial Respiration Is Promoted. Pathogens 2022; 11:pathogens11101168. [PMID: 36297225 PMCID: PMC9611969 DOI: 10.3390/pathogens11101168] [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: 09/02/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/19/2022] Open
Abstract
When exposed to a viral infection, the attacked cells promptly set up defense mechanisms. As part of the antiviral responses, the innate immune interferon pathway and associated interferon-stimulated genes notably allow the production of proteins bearing antiviral activity. Numerous viruses are able to evade the interferon response, highlighting the importance of controlling this pathway to ensure their efficient replication. Several viruses are also known to manipulate the metabolism of infected cells to optimize the availability of amino acids, nucleotides, and lipids. They then benefit from a reprogramming of the metabolism that favors glycolysis instead of mitochondrial respiration. Given the increasingly discussed crosstalk between metabolism and innate immunity, we wondered whether this switch from glycolysis to mitochondrial respiration would be beneficial or deleterious for an efficient antiviral response. We used a cell-based model of metabolic reprogramming. Interestingly, we showed that increased mitochondrial respiration was associated with an enhanced interferon response following polyriboinosinic:polyribocytidylic acid (poly:IC) stimulation. This suggests that during viral infection, the metabolic reprogramming towards glycolysis is also part of the virus’ strategies to inhibit the antiviral response.
Collapse
Affiliation(s)
- Grégorie Lebeau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
- Correspondence:
| | - Daed El Safadi
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| | - Aurélie Paulo-Ramos
- INSERM, UMR 1188 Diabète Athérothombose Réunion Océan Indien (DéTROI), Université de La Réunion, 97400 Saint-Denis, La Réunion, France
| | - Mathilde Hoareau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| | - Pascale Krejbich-Trotot
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| | - Florian Chouchou
- IRISSE Laboratory (EA4075), UFR SHE, University of La Réunion, 97430 Le Tampon, La Réunion, France
| | - Marjolaine Roche
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| | - Wildriss Viranaicken
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical, Plateforme Technologique CYROI, Université de La Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, 94791 Sainte Clotilde, La Réunion, France
| |
Collapse
|
87
|
Campos KB, Alomar AA, Eastmond BH, Obara MT, Alto BW. Brazilian Populations of Aedes aegypti Resistant to Pyriproxyfen Exhibit Lower Susceptibility to Infection with Zika Virus. Viruses 2022; 14:v14102198. [PMID: 36298753 PMCID: PMC9606930 DOI: 10.3390/v14102198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
Zika virus (ZIKV) infection has caused devastating consequences in Brazil as infections were associated with neurological complications in neonates. Aedes aegypti is the primary vector of ZIKV, and the evolution of insecticide resistance (IR) in this species can compromise control efforts. Although relative levels of phenotypic IR in mosquitoes can change considerably over time, its influence on vector competence for arboviruses is unclear. Pyriproxyfen (PPF)-resistant populations of Ae. aegypti were collected from five municipalities located in Northeast of Brazil, which demonstrated different resistance levels; low (Serrinha, Brumado), moderate (Juazeiro do Norte, Itabuna), and high (Quixadá). Experimental per os infection using ZIKV were performed with individuals from these populations and with an insecticide susceptible strain (Rockefeller) to determine their relative vector competence for ZIKV. Although all populations were competent to transmit ZIKV, mosquitoes derived from populations with moderate to high levels of IR exhibited similar or lower susceptibility to ZIKV infection than those from populations with low IR or the susceptible strain. These observations suggest an association between IR and arbovirus infection, which may be attributable to genetic hitchhiking. The use of PPF to control Brazilian Ae. aegypti may be associated with an indirect benefit of reduced susceptibility to infection, but no changes in disseminated infection and transmission of ZIKV among PPF-resistant phenotypes.
Collapse
Affiliation(s)
- Kauara Brito Campos
- Entomology and Nematology Department, Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, 200 9th SE, Vero Beach, FL 32962, USA
- Laboratório de Parasitologia Médica e Biologia de Vetores, Faculdade de Medicina, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Asa Norte, Brasília 70910-900, Brazil
- Coordenação Geral de Vigilância de Aboviroses, Secretaria de Vigilância em Saúde, Ministério da Saúde, Edifício PO 700, SRTV 702, Via W 5 Norte, Brasília 70723-040, Brazil
| | - Abdullah A. Alomar
- Entomology and Nematology Department, Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, 200 9th SE, Vero Beach, FL 32962, USA
| | - Bradley H. Eastmond
- Entomology and Nematology Department, Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, 200 9th SE, Vero Beach, FL 32962, USA
| | - Marcos Takashi Obara
- Laboratório de Parasitologia Médica e Biologia de Vetores, Faculdade de Medicina, Universidade de Brasília, Campus Universitário Darcy Ribeiro, Asa Norte, Brasília 70910-900, Brazil
| | - Barry W. Alto
- Entomology and Nematology Department, Florida Medical Entomology Laboratory, Institute of Food and Agricultural Sciences, University of Florida, 200 9th SE, Vero Beach, FL 32962, USA
- Correspondence:
| |
Collapse
|
88
|
Lee J, Park K, Kim J, Lee SH, Lee GY, Cho S, Kim HC, Klein TA, Kim JA, Choi J, Park J, Song DH, Gu SH, Yun H, Kim JE, Lee D, Hur GH, Jeong ST, Hwang IU, Kim WK, Song JW. Whole-genome sequencing and genetic diversity of severe fever with thrombocytopenia syndrome virus using multiplex PCR-based nanopore sequencing, Republic of Korea. PLoS Negl Trop Dis 2022; 16:e0010763. [PMID: 36094957 PMCID: PMC9499217 DOI: 10.1371/journal.pntd.0010763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 09/22/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022] Open
Abstract
Background
Whole-genome sequencing plays a critical role in the genomic epidemiology intended to improve understanding the spread of emerging viruses. Dabie bandavirus, causing severe fever with thrombocytopenia syndrome (SFTS), is a zoonotic tick-borne virus that poses a significant public health threat. We aimed to evaluate a novel amplicon-based nanopore sequencing tool to obtain whole-genome sequences of Dabie bandavirus, also known as SFTS virus (SFTSV), and investigate the molecular prevalence in wild ticks, Republic of Korea (ROK).
Principal findings
A total of 6,593 ticks were collected from Gyeonggi and Gangwon Provinces, ROK in 2019 and 2020. Quantitative polymerase chain reaction revealed the presence of SFSTV RNA in three Haemaphysalis longicornis ticks. Two SFTSV strains were isolated from H. longicornis captured from Pocheon and Cheorwon. Multiplex polymerase chain reaction-based nanopore sequencing provided nearly full-length tripartite genome sequences of SFTSV within one hour running. Phylogenetic and reassortment analyses were performed to infer evolutionary relationships among SFTSVs. Phylogenetic analysis grouped SFTSV Hl19-31-4 and Hl19-31-13 from Pocheon with sub-genotype B-1 in all segments. SFTSV Hl20-8 was found to be a genomic organization compatible with B-1 (for L segment) and B-2 (for M and S segments) sub-genotypes, indicating a natural reassortment between sub-genotypes.
Conclusion/Significance
Amplicon-based next-generation sequencing is a robust tool for whole-genome sequencing of SFTSV using the nanopore platform. The molecular prevalence and geographical distribution of SFTSV enhanced the phylogeographic map at high resolution for sophisticated prevention of emerging SFTS in endemic areas. Our findings provide important insights into the rapid whole-genome sequencing and genetic diversity for the genome-based diagnosis of SFTSV in the endemic outbreak.
Collapse
Affiliation(s)
- Jingyeong Lee
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kyungmin Park
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jongwoo Kim
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung-Ho Lee
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Geum-Young Lee
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seungchan Cho
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Heung-Chul Kim
- Force Health Protection and Preventive Medicine, Medical Department Activity-Korea/65th Medical Brigade, Unit 15281, United States of America
| | - Terry A. Klein
- Force Health Protection and Preventive Medicine, Medical Department Activity-Korea/65th Medical Brigade, Unit 15281, United States of America
| | - Jeong-Ah Kim
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Jeewan Choi
- Republic of Korea Armed Forces Medical Command, Seongnam, Republic of Korea
| | - Juwan Park
- The Fifth Preventive Medicine Unit of Republic of Korea Army, Pocheon, Republic of Korea
| | - Dong-Hyun Song
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Se Hun Gu
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Hyeongseok Yun
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Jung-Eun Kim
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Daesang Lee
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Gyeung Haeng Hur
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Seong Tae Jeong
- Chem-Bio Technology Center, Agency for Defense Development, Yuseong, Daejeon, Republic of Korea
| | - Il-Ung Hwang
- Department of Orthopaedic Surgery, Sheikh Khalifa Specialty Hospital, Seoul National University Hospital, Seoul, Republic of Korea
| | - Won-Keun Kim
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Institute of Medical Research, College of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jin-Won Song
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
89
|
Mendes Dos Santos MA, Dias LS, Ramirez Pavon JA, Viniski AE, Campos Souza CL, Pepato MA, Correa de Azevedo V, Teixeira Nunes MR, Slhessarenko RD. Regional mutations in CHIKV-ECSA genomes and detection of other viruses in the serum of acute febrile patients by a metagenomic approach in Mato Grosso, Central-Western Brazil, 2018. Virology 2022; 576:18-29. [PMID: 36126430 DOI: 10.1016/j.virol.2022.08.013] [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: 04/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/19/2022]
Abstract
Mato Grosso (MT) State is part of central western Brazil and has a tropical permissive environment that favors arbovirus outbreaks. A metagenomic approach was used to identify viral genomes in seven pools of serum from patients (n=65) with acute febrile disease. Seven chikungunya virus (CHIKV) genomes were determined, showing four amino acid changes found only in CHIKV genomes obtained in MT since 2018: nsP2:T31I, nsP3: A388V, E3:T201I and E3:H57R, in addition to other mutations in E1, nsP2 and nsP4. Six parvovirus B19 (B19V) genotype I genomes (4771-5131 nt) showed four aa alterations (NS1:N473D, R579Q; VP1:I716T; and 11 kDa:V44A) compared to most similar B19V from the USA. Coinfection between CHIKV and B19V was evidenced in 22/65 (33.8%) patients by RT‒PCR and PCR, respectively. Other viruses found in these pools include human pegivirus C, torque teno virus 3, an unclassified TTV and torque teno mini virus. Metagenomics represents a useful approach to detect viruses in the serum of acute febrile patients suspected of arbovirus disease.
Collapse
Affiliation(s)
- Marcelo Adriano Mendes Dos Santos
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil; Faculdade de Medicina, Universidade do Estado de Mato Grosso, Cáceres, MT, Brazil
| | - Lucas Silva Dias
- Curso de Graduação em Medicina, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | - Janeth Aracely Ramirez Pavon
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | - Ana Elisa Viniski
- Laboratório Central do Estado de Mato Grosso, Secretaria Estadoual de Saúde, Cuiabá, MT, Brazil
| | | | - Marco Andrey Pepato
- Laboratório Central do Estado de Mato Grosso, Secretaria Estadoual de Saúde, Cuiabá, MT, Brazil; Hospital Universitário Júlio Muller, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil
| | | | | | - Renata Dezengrini Slhessarenko
- Programa de Pós-Graduação em Ciências da Saúde, Faculdade de Medicina, Universidade Federal de Mato Grosso, Cuiabá, MT, Brazil.
| |
Collapse
|
90
|
Paixão ES, Fernandes QHRF, Cardim LL, Pescarini JM, Costa MCN, Falcão IR, Brickley EB, Santos AC, Portela Souza A, Carvalho-Sauer RDCO, Smeeth L, Rodrigues LC, Barreto ML, Teixeira MG. Socioeconomic risk markers of congenital Zika syndrome: a nationwide, registry-based study in Brazil. BMJ Glob Health 2022; 7:e009600. [PMID: 36175039 PMCID: PMC9528618 DOI: 10.1136/bmjgh-2022-009600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 08/29/2022] [Indexed: 11/04/2022] Open
Abstract
While it is well known that socioeconomic markers are associated with a higher risk of arbovirus infections, research on the relationship between socioeconomic factors and congenital Zika syndrome (CZS) remains limited. This study investigates the relationship between socioeconomic risk markers and live births with CZS in Brazil. We conducted a population-based study using data from all registered live births in Brazil (Live Births Information System) linked with the Public Health Event Record from 1 January 2015 to 31 December 2018. We used logistic regression models to estimate the OR and 95% CIs of CZS based on a three-level framework. In an analysis of 11 366 686 live births, of which 3353 had CZS, we observed that live births of self-identified black or mixed race/brown mothers (1.72 (95% CI 1.47 to 2.01) and 1.37 (95% CI 1.24 to 1.51)) were associated with a higher odds of CZS. Live births from single women compared with married women and those from women with less than 12 years of education compared with those with more than 12 years of education also had higher odds of CZS. In addition, live births following fewer prenatal care appointments had increased odds of CZS in the nationwide data. However, in the analyses conducted in the Northeast region (where the microcephaly epidemic started before the link with Zika virus was established and before preventive measures were known or disseminated), no statistical association was found between the number of prenatal care appointments and the odds of CZS. This study shows that live births of the most socially vulnerable women in Brazil had the greatest odds of CZS. This disproportionate distribution of risk places an even greater burden on already socioeconomically disadvantaged groups, and the lifelong disabilities caused by this syndrome may reinforce existing social and health inequalities.
Collapse
Affiliation(s)
- Enny S Paixão
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Qeren Hapuk R Ferreira Fernandes
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Luciana L Cardim
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Julia M Pescarini
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Ila R Falcão
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | - Elizabeth B Brickley
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Andreia Costa Santos
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - André Portela Souza
- School of Economics and Center for Applied Microeconomic Studies, Getulio Vargas Foundation, São Paulo, Brazil
| | | | - Liam Smeeth
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Laura C Rodrigues
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Mauricio L Barreto
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Brazil
| | | |
Collapse
|
91
|
Wang J, Gou QY, Luo GY, Hou X, Liang G, Shi M. Total RNA sequencing of Phlebotomus chinensis, a neglected vector in China, simultaneously revealed viral, bacterial, and eukaryotic microbes that are potentially pathogenic to humans. Emerg Microbes Infect 2022; 11:2080-2092. [PMID: 35916448 PMCID: PMC9448391 DOI: 10.1080/22221751.2022.2109516] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Phlebotomus chinensis sandfly is a neglected insect vector in China that is well-known for carrying Leishmania. Recent studies have expanded its pathogen repertoire with two novel arthropod-borne phleboviruses capable of infecting humans and animals. Despite these discoveries, our knowledge of the general pathogen diversity and overall microbiome composition of this vector species is still very limited. Here we carried out a meta-transcriptomics analysis that revealed the actively replicating/transcribing RNA viruses, DNA viruses, bacteria, and eukaryotic microbes, namely, the “total microbiome”, of several sandfly populations in China. Strikingly, “microbiome” made up 1.8% of total non-ribosomal RNA and comprised more than 87 species, among which 70 were novel, including divergent members of the genera Flavivirus and of the family Trypanosomatidae. Importantly, among these microbes we were able to reveal four distinguished types of human and/or mammalian pathogens, including two phleboviruses (hedi and wuxiang viruses), one novel Spotted fever group rickettsia, as well as a member of Leishmania donovani complex, among which hedi virus and Leishmania each had > 50% pool prevalence rate and relatively high abundance levels. Our study also showed the ubiquitous presence of an endosymbiont, namely Wolbachia, although no anti-viral or anti-pathogen effects were detected based on our data. In summary, our results uncovered the much un-explored diversity of microbes harboured by sandflies in China and demonstrated that high pathogen diversity and abundance are currently present in multiple populations, implying disease potential for exposed local human population or domestic animals.
Collapse
Affiliation(s)
- Jing Wang
- The Center for Infection & Immunity Study, School of Medicine, Shenzhen campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Qin-Yu Gou
- The Center for Infection & Immunity Study, School of Medicine, Shenzhen campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Geng-Yan Luo
- The Center for Infection & Immunity Study, School of Medicine, Shenzhen campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xin Hou
- The Center for Infection & Immunity Study, School of Medicine, Shenzhen campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Guodong Liang
- State Key Laboratory of Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Mang Shi
- The Center for Infection & Immunity Study, School of Medicine, Shenzhen campus of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
92
|
Dengue, Chikungunya, and Zika: Spatial and Temporal Distribution in Rio de Janeiro State, 2015–2019. Trop Med Infect Dis 2022; 7:tropicalmed7070141. [PMID: 35878153 PMCID: PMC9318038 DOI: 10.3390/tropicalmed7070141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
Simultaneous spatial circulation of urban arboviral diseases, such as dengue, chikungunya, and Zika, is a major challenge. In this ecological study of urban arboviruses performed from 2015 to 2019, we analyzed the spatial and temporal dynamics of these arboviruses in all 92 municipalities and nine health regions of Rio de Janeiro state. Annual cumulative incidences are presented for all three arboviruses throughout the study period. Spatial analyses of the three studied arboviruses showed distinct behaviors among municipalities and health regions. Co-circulation of the three arboviruses in the state and a heterogeneous spatiotemporal pattern was observed for each disease and region, with dengue having a higher annual incidence during the five years of the study, as well as two consecutive epidemic years in the state. The increase in transmission in different regions of the state in one year culminated in an epidemic in the state in the following year. A high annual cumulative incidence of chikungunya occurred in municipalities from 2017 to 2019 and of Zika only in 2016. Some municipalities with higher population densities showed higher incidences for some arboviruses and appeared to contribute to the dissemination to cities of lower demographic density and maintenance of these urban arboviruses. Thus, regions recording increased incidences of the three diseases in their territories for long periods should be considered municipal poles, as they initiated and sustained high transmission within their region.
Collapse
|
93
|
Lottini G, Baggiani M, Chesi G, D'Orsi B, Quaranta P, Lai M, Pancrazi L, Onorati M, Pistello M, Freer G, Costa M. Zika virus induces FOXG1 nuclear displacement and downregulation in human neural progenitors. Stem Cell Reports 2022; 17:1683-1698. [PMID: 35714598 PMCID: PMC9287670 DOI: 10.1016/j.stemcr.2022.05.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/25/2022] Open
Abstract
Congenital alterations in the levels of the transcription factor Forkhead box g1 (FOXG1) coding gene trigger "FOXG1 syndrome," a spectrum that recapitulates birth defects found in the "congenital Zika syndrome," such as microcephaly and other neurodevelopmental conditions. Here, we report that Zika virus (ZIKV) infection alters FOXG1 nuclear localization and causes its downregulation, thus impairing expression of genes involved in cell replication and apoptosis in several cell models, including human neural progenitor cells. Growth factors, such as EGF and FGF2, and Thr271 residue located in FOXG1 AKT domain, take part in the nuclear displacement and apoptosis protection, respectively. Finally, by progressive deletion of FOXG1 sequence, we identify the C-terminus and the residues 428-481 as critical domains. Collectively, our data suggest a causal mechanism by which ZIKV affects FOXG1, its target genes, cell cycle progression, and survival of human neural progenitors, thus contributing to microcephaly.
Collapse
Affiliation(s)
- Giulia Lottini
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy; Department of Medical Biotechnologies, University of Siena, Siena 53100, Italy
| | - Matteo Baggiani
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Giulia Chesi
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Beatrice D'Orsi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy
| | - Paola Quaranta
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Michele Lai
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Laura Pancrazi
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy
| | - Marco Onorati
- Unit of Cell and Developmental Biology, Department of Biology, University of Pisa, Pisa 56127, Italy
| | - Mauro Pistello
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Giulia Freer
- Centro Retrovirus, Department of Translational Research, University of Pisa, Pisa 56127, Italy
| | - Mario Costa
- Institute of Neuroscience, Italian National Research Council (CNR), Via Moruzzi, 1, Pisa 56124, Italy; Centro Pisano ricerca e implementazione clinica Flash Radiotherapy (CPFR@CISUP), Presidio S. Chiara, ed.18 via Roma, 67, Pisa 56126, Italy; Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Piazza dei Cavalieri, Pisa 56124, Italy.
| |
Collapse
|
94
|
Ward D, Gomes AR, Tetteh KKA, Sepúlveda N, Gomez LF, Campino S, Clark TG. Sero-epidemiological study of arbovirus infection following the 2015-2016 Zika virus outbreak in Cabo Verde. Sci Rep 2022; 12:11719. [PMID: 35810191 PMCID: PMC9271056 DOI: 10.1038/s41598-022-16115-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/05/2022] [Indexed: 11/09/2022] Open
Abstract
In November 2015, cases of Zika virus infection were recorded in Cabo Verde (Africa), originating from Brazil. The outbreak subsided after seven months with 7580 suspected cases. We performed a serological survey (n = 431) in Praia, the capital city, 3 months after transmission ceased. Serum samples were screened for arbovirus antibodies using ELISA techniques and revealed seroconverted individuals with Zika (10.9%), dengue (1-4) (12.5%), yellow fever (0.2%) and chikungunya (2.6%) infections. Zika seropositivity was predominantly observed amongst females (70%). Using a logistic model, risk factors for increased odds of Zika seropositivity included age, self-reported Zika infection, and dengue seropositivity. Serological data from Zika and dengue virus assays were strongly correlated (Spearman's rs = 0.80), which reduced when using a double antigen binding ELISA (Spearman's rs = 0.54). Overall, our work improves an understanding of how Zika and other arboviruses have spread throughout the Cabo Verde population. It also demonstrates the utility of serological assay formats for outbreak investigations.
Collapse
Affiliation(s)
- Daniel Ward
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | | | - Kevin K A Tetteh
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Nuno Sepúlveda
- Warsaw University of Technology, Warsaw, Poland
- Universidade de Lisboa, Lisbon, Portugal
| | | | - Susana Campino
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Taane G Clark
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK.
| |
Collapse
|
95
|
A fatal case of dengue hemorrhagic fever associated with dengue virus 4 (DENV-4) in Brazil: genomic and histopathological findings. Braz J Microbiol 2022; 53:1305-1312. [PMID: 35779208 DOI: 10.1007/s42770-022-00784-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/10/2022] [Indexed: 11/02/2022] Open
Abstract
Dengue infection is the most prevalent arthropod-borne viral disease in subtropical and tropical regions, whose primary vector is Aedes aegypti mosquitoes. The mechanisms of dengue virus (DENV) pathogenesis are little understood because we have no good disease models. Only humans develop symptoms (dengue fever, DF, or dengue hemorrhagic fever, DHF) and research has been limited to studies involving patients. Samples from serum, brain, cerebellum, heart, lungs, liver, and kidneys from a 13-year-old male patient that died with hemorrhagic manifestations were sent for differential diagnosis at Adolfo Lutz, using both classical virological methods (RT-qPCR, virus isolation, ELISA, and hemagglutination inhibition test) and immunohistochemistry (IHQ). A DENV serotype 4 was detected by a DENV multiplex RT-qPCR, and the C6/36 cell supernatant was used for NGS using Minion. Lesions were described in the heart, liver, lung, and kidney with positive IHQ in endothelial cells of the brain, cerebellum, heart, and kidney, and also in hepatocytes and Kuppfer cells. A whole genome was obtained, revealing a DENV-4 genotype II, with no evidence of secondary dengue infection.
Collapse
|
96
|
Jamieson T, Rivera JJC. Our issue or their issue? Media coverage and framing of the Zika virus epidemic. DISASTERS 2022; 46:677-699. [PMID: 34197015 DOI: 10.1111/disa.12497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
How does the news media respond to health emergencies abroad? Between 2015 and 2018, Zika virus spread rapidly throughout Latin America before arriving in the continental United States. Despite the risks to adults and newborns, it is unclear how media coverage developed and framed the threat for its audience. In this paper, we argue that while the frequency of coverage was responsive to infections, its content failed to promote proactive health behaviour. To assess these claims, we analyse each of 442 articles dealing with Zika virus published by The New York Times from 2015-18. We find that the amount of coverage reflected infections but did not change once the disease emerged in the US. Furthermore, content analysis using Linguistic Inquiry and Word Count software reveals that coverage emphasised differences between communities (those affected and those at home) and that present and past time orientations dominated coverage as opposed to future time orientations.
Collapse
Affiliation(s)
- Thomas Jamieson
- Assistant Professor, School of Public Administration, University of Nebraska at Omaha, United States
| | | |
Collapse
|
97
|
Gupta AK, Kumar M. Benchmarking and Assessment of Eight De Novo Genome Assemblers on Viral Next-Generation Sequencing Data, Including the SARS-CoV-2. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:372-381. [PMID: 35759429 DOI: 10.1089/omi.2022.0042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Viral genomics has become crucial in clinical diagnostics and ecology, not to mention to stem the COVID-19 pandemic. Whole-genome sequencing (WGS) is pivotal in gaining an improved understanding of viral evolution, genomic epidemiology, infectious outbreaks, pathobiology, clinical management, and vaccine development. Genome assembly is one of the crucial steps in WGS data analyses. A series of different assemblers has been developed with the advent of high-throughput next-generation sequencing (NGS). Various studies have reported the evaluation of these assembly tools on distinct datasets; however, these lack data from viral origin. In this study, we performed a comparative evaluation and benchmarking of eight de novo assemblers: SOAPdenovo, Velvet, assembly by short sequences (ABySS), iterative De Bruijn graph assembler (IDBA), SPAdes, Edena, iterative virus assembler, and VICUNA on the viral NGS data from distinct Illumina (GAIIx, Hiseq, Miseq, and Nextseq) platforms. WGS data of diverse viruses, that is, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), dengue virus 3, human immunodeficiency virus 1, hepatitis B virus, human herpesvirus 8, human papillomavirus 16, rhinovirus A, and West Nile virus, were utilized to assess these assemblers. Performance metrics such as genome fraction recovery, assembly lengths, NG50, N50, contig length, contig numbers, mismatches, and misassemblies were analyzed. Overall, three assemblers, that is, SPAdes, IDBA, and ABySS, performed consistently well, including for genome assembly of SARS-CoV-2. These assembly methods should be considered and recommended for future studies of viruses. The study also suggests that implementing two or more assembly approaches should be considered in viral NGS studies, especially in clinical settings. Taken together, the benchmarking of eight de novo genome assemblers reported in this study can inform future public health and ecology research concerning the viruses, the COVID-19 pandemic, and viral outbreaks.
Collapse
Affiliation(s)
- Amit Kumar Gupta
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Chandigarh, India
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
98
|
Eberle RJ, Olivier DS, Amaral MS, Pacca CC, Nogueira ML, Arni RK, Willbold D, Coronado MA. Riboflavin, a Potent Neuroprotective Vitamin: Focus on Flavivirus and Alphavirus Proteases. Microorganisms 2022; 10:1331. [PMID: 35889050 PMCID: PMC9315535 DOI: 10.3390/microorganisms10071331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 12/01/2022] Open
Abstract
Several neurotropic viruses are members of the flavivirus and alphavirus families. Infections caused by these viruses may cause long-term neurological sequelae in humans. The continuous emergence of infections caused by viruses around the world, such as the chikungunya virus (CHIKV) (Alphavirus genus), the zika virus (ZIKV) and the yellow fever virus (YFV) (both of the Flavivirus genus), warrants the development of new strategies to combat them. Our study demonstrates the inhibitory potential of the water-soluble vitamin riboflavin against NS2B/NS3pro of ZIKV and YFV and nsP2pro of CHIKV. Riboflavin presents a competitive inhibition mode with IC50 values in the medium µM range of 79.4 ± 5.0 µM for ZIKV NS2B/NS3pro and 45.7 ± 2.9 μM for YFV NS2B/NS3pro. Against CHIKV nsP2pro, the vitamin showed a very strong effect (93 ± 5.7 nM). The determined dissociation constants (KD) are significantly below the threshold value of 30 µM. The ligand binding increases the thermal stability between 4 °C and 8 °C. Unexpectedly, riboflavin showed inhibiting activity against another viral protein; the molecule was also able to inhibit the viral entry of CHIKV. Molecular dynamics simulations indicated great stability of riboflavin in the protease active site, which validates the repurposing of riboflavin as a promising molecule in drug development against the viruses presented here.
Collapse
Affiliation(s)
- Raphael J. Eberle
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
| | - Danilo S. Olivier
- Center of Integrated Sciences, Campus Cimba, Federal University of Tocantins, Araguaína 77824-838, TO, Brazil;
| | - Marcos S. Amaral
- Institute of Physics, Federal University of Mato Grosso do Sul, Campo Grande 79070-900, MS, Brazil;
| | - Carolina C. Pacca
- Instituto Superior de Educação Ceres, FACERES Medical School, São José do Rio Preto 15090-305, SP, Brazil;
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto-FAMERP, São José do Rio Preto 15090-000, SP, Brazil;
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Departamento de Doenças Dermatológicas, Infecciosas e Parasitárias, Faculdade de Medicina de São José do Rio Preto-FAMERP, São José do Rio Preto 15090-000, SP, Brazil;
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Raghuvir K. Arni
- Multiuser Center for Biomolecular Innovation, Department of Physics, IBILCE, São Paulo State University, São Jose do Rio Preto 15054-000, SP, Brazil;
| | - Dieter Willbold
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße, 40225 Düsseldorf, Germany
- JuStruct: Jülich Centre for Structural Biology, Forchungszentrum Jülich, 52428 Jülich, Germany
| | - Monika A. Coronado
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany;
| |
Collapse
|
99
|
Gao Y, Tai W, Wang X, Jiang S, Debnath AK, Du L, Chen S. A gossypol derivative effectively protects against Zika and dengue virus infection without toxicity. BMC Biol 2022; 20:143. [PMID: 35706035 PMCID: PMC9202104 DOI: 10.1186/s12915-022-01344-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 06/07/2022] [Indexed: 11/28/2022] Open
Abstract
Background Zika virus (ZIKV) and dengue virus (DENV) cause microcephaly and dengue hemorrhagic fever, respectively, leading to severe problems. No effective antiviral agents are approved against infections of these flaviviruses, calling for the need to develop potent therapeutics. We previously identified gossypol as an effective inhibitor against ZIKV and DENV infections, but this compound is toxic and not suitable for in vivo treatment. Results In this study, we showed that gossypol derivative ST087010 exhibited potent and broad-spectrum in vitro inhibitory activity against infections of at least ten ZIKV strains isolated from different hosts, time periods, and countries, as well as DENV-1-4 serotypes, and significantly reduced cytotoxicity compared to gossypol. It presented broad-spectrum in vivo protective efficacy, protecting ZIKV-infected Ifnar1−/− mice from lethal challenge, with increased survival and reduced weight loss. Ifnar1−/− mice treated with this gossypol derivative decreased viral titers in various tissues, including the brain and testis, after infection with ZIKV at different human isolates. Moreover, ST087010 potently blocked ZIKV vertical transmission in pregnant Ifnar1−/− mice, preventing ZIKV-caused fetal death, and it was safe for pregnant mice and their pups. It also protected DENV-2-challenged Ifnar1−/− mice against viral replication by reducing the viral titers in the brain, kidney, heart, and sera. Conclusions Overall, our data indicate the potential for further development of this gossypol derivative as an effective and safe broad-spectrum therapeutic agent to treat ZIKV and DENV diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01344-w.
Collapse
Affiliation(s)
- Yaning Gao
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.,Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Wanbo Tai
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Xinyi Wang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.,Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Asim K Debnath
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, 10065, USA.
| | - Shizhong Chen
- Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
100
|
Aggio JB, Porto BN, Duarte dos Santos CN, Mosimann ALP, Wowk PF. Human Neutrophils Present Mild Activation by Zika Virus But Reduce the Infection of Susceptible Cells. Front Immunol 2022; 13:784443. [PMID: 35747137 PMCID: PMC9210994 DOI: 10.3389/fimmu.2022.784443] [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: 09/27/2021] [Accepted: 05/02/2022] [Indexed: 11/16/2022] Open
Abstract
The emergence of the Zika virus (ZIKV) has highlighted the need for a deeper understanding of virus-host interactions in order to pave the way for the development of antiviral therapies. The present work aimed to address the response of neutrophils during ZIKV infection. Neutrophils are important effector cells in innate immunity implicated in the host’s response to neurotropic arboviruses. Our results indicate that human neutrophils were not permissive to Asian or African ZIKV strain replication. In fact, after stimulation with ZIKV, neutrophils were mild primed against the virus as evaluated through CD11b and CD62L modulation, secretion of inflammatory cytokines and granule content, production of reactive oxygen species, and neutrophil extracellular traps formation. Overall, neutrophils did not affect ZIKV infectivity. Moreover, in vitro ZIKV infection of primary innate immune cells did not trigger neutrophil migration. However, neutrophils co-cultured with ZIKV susceptible cell lineages resulted in lower cell infection frequencies, possibly due to cell-to-cell contact. In vivo, neutrophil depletion in immunocompetent mice did not affect ZIKV spreading to the draining lymph nodes. The data suggest that human neutrophils do not play an antiviral role against ZIKV per se, but these cells might participate in an infected environment shaping the ZIKV infection in other target cells.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
| | - Bárbara Nery Porto
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB, Canada
- Biology of Breathing Group, Children’s Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | | | - Ana Luiza Pamplona Mosimann
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
- *Correspondence: Pryscilla Fanini Wowk, ; Ana Luiza Pamplona Mosimann,
| | - Pryscilla Fanini Wowk
- Laboratório de Virologia Molecular, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ), Curitiba, Brazil
- *Correspondence: Pryscilla Fanini Wowk, ; Ana Luiza Pamplona Mosimann,
| |
Collapse
|