1
|
Cong H, Han X, Lv M, Chang Y, Wang X, Lei R. Integrated duplex reverse transcription-recombinase aided amplification (RT-RAA) and lateral flow assay (LFA) for rapid simultaneous detection of Zika virus and Japanese encephalitis virus in single reaction format. Talanta 2025; 294:128195. [PMID: 40315800 DOI: 10.1016/j.talanta.2025.128195] [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: 01/27/2025] [Revised: 04/06/2025] [Accepted: 04/20/2025] [Indexed: 05/04/2025]
Abstract
Zika virus (ZIKV) and Japanese encephalitis virus (JEV), two consequential mosquito-borne flaviviruses, induce severe systemic and neurotropic-encephalitic pathologies with overlapping symptoms profiles, complicating differential diagnosis in co-endemic regions. To address this critical challenge, we developed a dual reverse-transcription recombinase-aided amplification (RT-RAA) coupled with duplex lateral flow assay (LFA) platform for rapid, equipment-free co-detection of ZIKV and JEV in a single reaction tube. The assay achieved isothermal amplification at 39 °C within 40 min using a field-deployable incubator. The limit of detection (LOD) reached as low as 8.5 copies ZIKV RNA, and 1.1 copies JEV RNA in single pathogen detection, while 110 copies JEV RNA in co-detection mode. This technological advance bridges the critical sensitivity gap between lab-based PCR and conventional rapid tests, enabling first-line healthcare responders to conduct precision diagnostics in non-laboratory settings from rural clinics to mobile outbreak response units.
Collapse
Affiliation(s)
- Haolong Cong
- Center for Biosafety, Chinese Academy of Inspection and Quarantine, Sanya, 572024, China; Chinese Academy of Quality and Inspection & Testing, Beijing, 100176, China
| | - Xiaodong Han
- College of Life Sciences, Inner Mongolia Agriculture University, Hohhot, Inner Mongolia, China
| | - Mengyuan Lv
- School of Life and Health, Dalian University, Dalian, 116622, China
| | - Yutong Chang
- Chinese Academy of Quality and Inspection & Testing, Beijing, 100176, China
| | - Xinyi Wang
- School of Life and Health, Dalian University, Dalian, 116622, China.
| | - Rong Lei
- Center for Biosafety, Chinese Academy of Inspection and Quarantine, Sanya, 572024, China; Chinese Academy of Quality and Inspection & Testing, Beijing, 100176, China.
| |
Collapse
|
2
|
Van der Hoek KH, Jankovic-Karasoulos T, McCullough D, Coldbeck-Shackley RC, Eyre NS, Roberts CT, Beard MR. The first trimester human placenta responds to Zika virus infection inducing an interferon (IFN) and antiviral interferon stimulated gene (ISG) response. Virol J 2025; 22:108. [PMID: 40253335 PMCID: PMC12008946 DOI: 10.1186/s12985-025-02729-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/07/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Zika virus (ZIKV) is a positive-strand RNA virus of the Flaviviridae family. Maternal ZIKV infection during pregnancy can spread to the placenta and fetus causing severe neurological defects and infants born with microcephaly. Here, we investigated ZIKV infection and the cellular innate antiviral immune response in first trimester human placental explant cultures and isolated primary villus cytotrophoblasts (CTBs). METHODS Placentas were obtained with informed consent from women undergoing elective pregnancy termination and either cultured as placental explants or used to isolate primary CTBs. Explants and CTBs were both infected with ZIKV (PRVABC59), and samples evaluated for infection by qRT-PCR, viral plaque and ELISA assays, and immunohistochemical or immunocytochemical staining. RESULTS We demonstrate robust infection and production of ZIKV in placental explant and CTB cultures. Both displayed delayed upregulation of interferons (IFN), most notably IFNβ and IFNλ2/3, and a panel of interferon stimulated genes (ISG) (IFI6, IFIT1, IFIT2, IFITM1, ISG15, MX1, RSAD). Stimulation of explants and CTBs with the dsRNA mimic poly(I: C), caused immediate IFN and ISG upregulation, demonstrating the first trimester placenta is innate immune competent. This suggests that either ZIKV blocks the early innate response, or the placental response is inherently hindered. CONCLUSION Together these data show that first trimester placenta is susceptible to ZIKV infection which induces a delayed type III IFN antiviral response. This delay likely creates an environment favourable to ZIKV replication and dissemination across the early gestation placenta to fetal tissue, causing pathologies associated with congenital ZIKV syndrome.
Collapse
Affiliation(s)
- Kylie H Van der Hoek
- Research Centre for Infectious Diseases, The University of Adelaide, Adelaide, SA, 5005, Australia.
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia.
- Department of Molecular and Biomedical Science and Research Centre for Infectious Diseases, University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Tanja Jankovic-Karasoulos
- University of Adelaide, The Robinson Research Institute, Adelaide, SA, 5005, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, 5005, Australia
| | - Dylan McCullough
- University of Adelaide, The Robinson Research Institute, Adelaide, SA, 5005, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, 5005, Australia
| | - Rosa C Coldbeck-Shackley
- Research Centre for Infectious Diseases, The University of Adelaide, Adelaide, SA, 5005, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Nicholas S Eyre
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, 5005, Australia
| | - Claire T Roberts
- University of Adelaide, The Robinson Research Institute, Adelaide, SA, 5005, Australia
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, SA, 5005, Australia
| | - Michael R Beard
- Research Centre for Infectious Diseases, The University of Adelaide, Adelaide, SA, 5005, Australia
- School of Biological Sciences, The University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
3
|
Pang M, Sun XZ, He T, Yang H, Chen J. Clinical Manifestation of Arboviruses in Paediatrics. Rev Med Virol 2025; 35:e70016. [PMID: 39934924 DOI: 10.1002/rmv.70016] [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: 12/11/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Arboviral infections in paediatric populations present unique challenges due to distinct pathophysiological mechanisms influenced by developmental and immunological differences. Commonly implicated arboviruses include dengue virus (DENV), Zika virus (ZIKV), chikungunya virus (CHIKV), West Nile virus (WNV), and yellow fever virus (YFV). These viruses exhibit specific tropisms, targeting organs such as the central nervous system (CNS), liver, and vasculature. Immune responses in children, characterised by an underdeveloped adaptive system and enhanced innate immunity, can exacerbate inflammation and increase susceptibility to severe outcomes such as dengue hemorrhagic fever (DHF), congenital Zika syndrome (CZS), and neuroinvasive complications. Maternal antibodies, antibody-dependent enhancement (ADE), and immature barriers, such as the blood-brain barrier, further contribute to disease severity. This review highlights the virological and immunological nuances of arboviral pathophysiology in paediatric patients, emphasising the need for age-specific diagnostic, therapeutic, and preventive strategies to mitigate the burden of these infections.
Collapse
Affiliation(s)
- Mu Pang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chineses Medicine Hospital), Shenzhen, China
| | - Xi-Zhe Sun
- Research Center for Drug Safety Evalution of Hainan Province, Hainan Medical University, Haikou, China
| | - Ting He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chineses Medicine Hospital), Shenzhen, China
| | - Hao Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chineses Medicine Hospital), Shenzhen, China
| | - Jun Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine (Shenzhen Traditional Chineses Medicine Hospital), Shenzhen, China
| |
Collapse
|
4
|
dos Santos ALS, Rosolen BB, Ferreira FC, Chiancone IS, Pereira SS, Pontes KFM, Traina E, Werner H, Granese R, Araujo Júnior E. Intrauterine Zika Virus Infection: An Overview of the Current Findings. J Pers Med 2025; 15:98. [PMID: 40137414 PMCID: PMC11943202 DOI: 10.3390/jpm15030098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus of the family Flaviviridae. The association between ZIKV and microcephaly was first described in Brazil in 2015. The risk of vertical transmission occurs in pregnant women with or without symptoms, and the risk of malformation appears to be worse when infection occurs in the first and second trimesters of pregnancy. The rate of vertical transmission varies from 26 to 65%, and not all fetuses develop malformations. The incidence of malformations resulting from transmission is uncertain, ranging from 6-8% in the US to 40% in Brazil. Congenital ZIKV syndrome is a set of clinical manifestations that can affect the fetus of a mother infected with ZIKV. The manifestations are broad and nonspecific, including microcephaly, subcortical calcifications, ocular changes, congenital contractures, early hypertension, and pyramidal and extrapyramidal signs. Other findings such as growth restriction and fetal miscarriage/death may also occur. Our aim in this article is to review the literature on mosquito transmission, clinical presentation, serologic diagnosis, intrauterine transmission, pre- and postnatal imaging diagnostic findings, and short- and long-term follow-up.
Collapse
Affiliation(s)
- Ana Luiza Soares dos Santos
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Beatriz Bussi Rosolen
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Fernanda Curvelo Ferreira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Isabella Samões Chiancone
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Stefany Silva Pereira
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
| | - Karina Felippe Monezi Pontes
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
- Service of Gynecology and Obstetrics, Ipiranga Hospital, São Paulo 04262-000, SP, Brazil
| | - Evelyn Traina
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| | - Heron Werner
- Department of Fetal Medicine, Biodesign Laboratory DASA/PUC, Rio de Janeiro 22453-900, RJ, Brazil;
| | - Roberta Granese
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, “G. Martino” University Hospital, 98100 Messina, Italy
| | - Edward Araujo Júnior
- Discipline of Woman Health, Municipal University of São Caetano do Sul (USCS), São Caetano do Sul 09521-160, SP, Brazil; (A.L.S.d.S.); (B.B.R.); (F.C.F.); (I.S.C.); (S.S.P.); (E.A.J.)
- Department of Obstetrics, Paulista School of Medicine, Federal University of São Paulo (EPM-UNIFESP), São Paulo 04023-062, SP, Brazil; (K.F.M.P.); (E.T.)
| |
Collapse
|
5
|
Ni X, Richardson RB, Godoy AS, Ferla MP, Kikawa C, Scheen J, Hannon WW, Capkin E, Lahav N, Balcomb BH, Marples PG, Fairhead M, Wang S, Williams EP, Tomlinson CWE, Aschenbrenner JC, Lithgo RM, Winokan M, Giroud C, Chandran AV, Walsh M, Thompson W, Bloom JD, Barr H, Kirkegaard K, Koekemoer L, Fearon D, Evans MJ, von Delft F. Crystallographic fragment screening and deep mutational scanning of Zika virus NS2B-NS3 protease enable development of resistance-resilient inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.29.591502. [PMID: 38746305 PMCID: PMC11092485 DOI: 10.1101/2024.04.29.591502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The Zika viral protease NS2B-NS3 is essential for the cleavage of viral polyprotein precursor into individual structural and non-structural (NS) proteins and is therefore an attractive drug target. Generation of a robust crystal system of co-expressed NS2B-NS3 protease has enabled us to perform a crystallographic fragment screening campaign with 1076 fragments. 47 fragments with diverse scaffolds were identified to bind in the active site of the protease, with another 6 fragments observed in a potential allosteric site. To identify binding sites that are intolerant to mutation and thus suppress the outgrowth of viruses resistant to inhibitors developed from bound fragments, we performed deep mutational scanning of NS2B-NS3 protease. Merging fragment hits yields an extensive set of 'mergers', defined as synthetically accessible compounds that recapitulate constellations of observed fragment-protein interactions. In addition, the highly sociable fragment hits enable rapid exploration of chemical space via algorithmic calculation and thus yield diverse possible starting points that maximally explore the binding opportunities to NS2B-NS3 protease, facilitating its resistance-resilient antiviral development.
Collapse
Affiliation(s)
- Xiaomin Ni
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - R Blake Richardson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andre Schutzer Godoy
- São Carlos Institute of Physics, University of São Paulo, Av. João Dagnone, 1100 - Jardim Santa Angelina, São Carlos, 13563-120, Brazil
| | - Matteo P Ferla
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Caroline Kikawa
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Division of Basic Sciences, Fred Hutch Cancer Center, Seattle, WA, USA
| | - Jenke Scheen
- Open Molecular Software Foundation, Davis, CA 95618, USA
| | - William W Hannon
- Division of Basic Sciences, Computational Biology Program, and Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Eda Capkin
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
| | - Noa Lahav
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science
| | - Blake H Balcomb
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Peter G Marples
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Michael Fairhead
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Siyi Wang
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Eleanor P Williams
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Charlie W E Tomlinson
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Jasmin Cara Aschenbrenner
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Ryan M Lithgo
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Max Winokan
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Charline Giroud
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Anu V Chandran
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Martin Walsh
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Warren Thompson
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Jesse D Bloom
- Division of Basic Sciences, Fred Hutch Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, Washington, USA
| | - Haim Barr
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science
| | - Karla Kirkegaard
- Department of Genetics, Stanford University, Palo Alto, California, USA
- Department of Microbiology and Immunology, Stanford University, Palo Alto, California, USA
| | - Lizbé Koekemoer
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
| | - Daren Fearon
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| | - Matthew J Evans
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Frank von Delft
- Centre for Medicines Discovery, NDM research Building, Roosevelt Dr, Headington, Oxford OX3 7FZ, United Kingdom
- Diamond Light Source Ltd, Harwell Science and Innovation Campus, Didcot, Oxfordshire, OX11 0DE, United Kingdom
- Research Complex at Harwell, Harwell Science and Innovation Campus, Didcot, UK
| |
Collapse
|
6
|
von Delft F, Ni X, Richardson R, Godoy A, Ferla M, Kikawa C, Scheen J, Hannon W, Capkin E, Lahav N, Balcomb B, Marples P, Fairhead M, Wang S, Williams E, Tomlinson C, Aschenbrenner J, Lithgo R, Winokan M, Giroud C, Chandran A, Walsh M, Thompson W, Bloom J, Barr H, Kirkegaard K, Koekemoer L, Fearon D, Evans M. Crystallographic fragment screening and deep mutational scanning of Zika virus NS2B-NS3 protease enable development of resistance-resilient inhibitors. RESEARCH SQUARE 2025:rs.3.rs-5876218. [PMID: 39989958 PMCID: PMC11844641 DOI: 10.21203/rs.3.rs-5876218/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The Zika viral protease NS2B-NS3 is essential for the cleavage of viral polyprotein precursor into individual structural and non-structural (NS) proteins and is therefore an attractive drug target. Generation of a robust crystal system of co-expressed NS2B-NS3 protease has enabled us to perform a crystallographic fragment screening campaign with 1076 fragments. 47 fragments with diverse scaffolds were identified to bind in the active site of the protease, with another 6 fragments observed in a potential allosteric site. To identify binding sites that are intolerant to mutation and thus suppress the outgrowth of viruses resistant to inhibitors developed from bound fragments, we performed deep mutational scanning of NS2B-NS3 protease. Merging fragment hits yields an extensive set of 'mergers', defined as synthetically accessible compounds that recapitulate constellations of observed fragment-protein interactions. In addition, the highly sociable fragment hits enable rapid exploration of chemical space via algorithmic calculation and thus yield diverse possible starting points that maximally explore the binding opportunities to NS2B-NS3 protease, facilitating its resistance-resilient antiviral development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Noa Lahav
- The Wohl Drug Discovery Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Torii S, Lord JS, Lavina M, Prot M, Lecuyer A, Diagne CT, Faye O, Faye O, Sall AA, Bonsall MB, Simon-Lorière E, Montagutelli X, Lambrechts L. Polygenic viral factors enable efficient mosquito-borne transmission of African Zika virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634482. [PMID: 39896559 PMCID: PMC11785240 DOI: 10.1101/2025.01.23.634482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus primarily transmitted among humans by Aedes aegypti. Over the past two decades, it has caused significant outbreaks associated with birth defects and neurological disorders. Phylogenetically, ZIKV consists of two main genotypes referred to as the African and Asian lineages, each exhibiting distinct biological properties. African lineage strains are transmitted more efficiently by mosquitoes, but pinpointing the genetic basis of this difference has remained challenging. Here, we address this question by comparing recent African and Asian strains using chimeric viruses, in which segments of the parental genomes are swapped. Our results show that the structural genes from the African strain enhance viral internalization, while the non-structural genes improve genome replication and infectious particle production in mosquito cells. In vivo mosquito transmission is most significantly influenced by the structural genes, although no single viral gene alone determines this effect. Additionally, we develop a stochastic model of in vivo viral dynamics in mosquitoes that mirrors the observed patterns, suggesting that the primary difference between the African and Asian strains lies in their ability to traverse the mosquito salivary glands. Overall, our findings suggest that the polygenic nature of ZIKV transmissibility has prevented Asian lineage strains from achieving the same epidemic potential as African lineage strains, underscoring the importance of lineage-specific adaptive landscapes in shaping ZIKV evolution and emergence.
Collapse
Affiliation(s)
- Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Jennifer S. Lord
- Department of Vector Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Morgane Lavina
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Matthieu Prot
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Alicia Lecuyer
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Cheikh T. Diagne
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Oumar Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Ousmane Faye
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | - Amadou A. Sall
- Arbovirus and Viral Hemorrhagic Fevers Unit, Institut Pasteur de Dakar, Dakar, Senegal
| | | | - Etienne Simon-Lorière
- Institut Pasteur, Université Paris Cité, Paris, Evolutionary Genomics of RNA Viruses Unit, Paris France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| |
Collapse
|
8
|
Morrey JD, Siddharthan V, Wang H, Oliveira ALR, Susuki K, Kaundal R, Freeman SM, Thomas AJ, Duhan N, Corry NG. Identification of candidate genes involved in Zika virus-induced reversible paralysis of mice. Sci Rep 2025; 15:2926. [PMID: 39848964 PMCID: PMC11757732 DOI: 10.1038/s41598-025-86475-0] [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: 08/30/2024] [Accepted: 01/10/2025] [Indexed: 01/25/2025] Open
Abstract
Zika virus (ZIKV) causes a variety of peripheral and central nervous system complications leading to neurological symptoms such as limb weakness. We used a mouse model to identify candidate genes potentially involved in causation or recovery from ZIKV-induced acute flaccid paralysis. Using Zikv and Chat chromogenic and fluorescence in situ RNA hybridization, electron microscopy, immunohistochemistry, and ZIKV RT-qPCR, we determined that some paralyzed mice had infected motor neurons, but motor neurons are not reduced in number and the infection was not present in all paralyzed mice; hence infection of motor neurons were not strongly correlated with paralysis. Consequently, paralysis was probably caused by by-stander effects. To address this, we performed bioinformatics analysis on spinal cord RNA to identify 2058 differentially expressed genes (DEGs) that were altered during paralysis and then normalized after paralysis. Of these "biphasic" DEGs, 951 were up-regulated and 1107 were down-regulated during paralysis, followed by recovery. To refine the search for candidate DEGs we used gene ontology analysis and RT-qPCR to select 3 DEGs that could be involved with the node of Ranvier function and 5 DEGs that could be involved with synaptic function. Among these, SparcL1:Sparc DEG ratios were identified to be inversely correlated with ZIKV-induced paralysis, which is consistent with the known function of SPARC protein to antagonize the synaptogenesis of SPARCL1. Ank3, Sptbn1, and Epb41l3 affecting the structures at and near the nodes of Ranvier were significantly downregulated during ZIKV-induced paralysis. The primary contribution is the identification of 8 candidate genes that may be involved in the causation or recovery of ZIKV-induced paralysis.
Collapse
Affiliation(s)
- John D Morrey
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84321-5600, USA.
| | - Venkatraman Siddharthan
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84321-5600, USA
| | - Hong Wang
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84321-5600, USA
| | | | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435-0001, USA
| | - Rakesh Kaundal
- Bioinformatics Facility, Center for Integrated BioSystems, Department of Plants, Soils, and Climate, College of Agriculture and Applied Sciences, Logan, UT, 84322, USA
- Department of Computer Science, College of Science, Logan, UT, 84322, USA
| | - Sara M Freeman
- Department of Biology, Utah State University, Logan, UT, 84322, USA
| | - Aaron J Thomas
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84322, USA
| | - Naveen Duhan
- Department of Plants, Soils, and Climate, College of Agriculture and Applied Sciences, Logan, UT, 84322, USA
| | - Nathan G Corry
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT, 84321-5600, USA
| |
Collapse
|
9
|
Rahman T, Bappi MSH, Hossain TJ. Prodigiosin Demonstrates Promising Antiviral Activity Against Dengue Virus and Zika Virus in In-silico Study. ANALYTICAL SCIENCE ADVANCES 2024; 5:e202400039. [PMID: 39660345 PMCID: PMC11627182 DOI: 10.1002/ansa.202400039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 12/12/2024]
Abstract
Dengue (DENV) and Zika virus (ZIKV), transmitted by Aedes mosquitoes, pose significant public health challenges. Effective treatments for these viruses remain elusive, highlighting the urgent need for new efficient antiviral therapies. This study explores prodigiosin, a microbial tripyrrole pigment, as an antiviral agent against both DENV and ZIKV employing advanced analytical approaches which integrate molecular docking, CASTp 3.0 validation and molecular dynamics (MD) simulations providing insights into molecular interactions at an atomic level. Prodigiosin exhibited favourable drug-likeness properties, meeting Lipinski's rule of five and demonstrating optimal physicochemical and pharmacokinetic characteristics according to Ghose's, Veber's, Egan's and Muegge's filters, essential for oral bioavailability. Absorption, Distribution, Metabolism, Excretion, and Toxicity profiling indicated high intestinal absorption, minimal risk for drug-drug interactions and a low toxicity profile, with no AMES toxicity, hepatotoxicity, or skin sensitization. Molecular docking revealed prodigiosin's strong binding affinities to NS5 methyltransferases of both DENV (-7.6 kcal/mol) and ZIKV (-7.7 kcal/mol) viruses, suggesting potential disruption of viral replication. Notably, prodigiosin's binding affinities were comparable to ribavirin-5'-triphosphate and chloroquine, known inhibitors of DENV and ZIKV, respectively. MD simulations confirmed stable and specific interactions with prodigiosin with low root-mean-square deviation values. Additional analyses, including root-mean-square fluctuation, radius of gyration and solvent-accessible surface area, indicated compact and stable complexes. These multi-parametric in-silico analytical strategies provide a novel perspective of prodigiosin as an antiviral agent, demonstrating its drug interactions at the molecular level. These promising results suggest that prodigiosin could serve as a broad-spectrum antiviral agent against both DENV and ZIKV, warranting further experimental validation for therapeutic development against flaviviral infections.
Collapse
Affiliation(s)
- Tanjilur Rahman
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChattogramBangladesh
| | - Mohammed Sajjad Hossain Bappi
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChattogramBangladesh
- Biotechnology, Informatics and Genomics (BIG) Unit, Laboratory for HealthOmics and Pathway Exploration (HOPE Lab)ChattogramBangladesh
| | - Tanim Jabid Hossain
- Department of Biochemistry and Molecular BiologyUniversity of ChittagongChattogramBangladesh
- Biotechnology, Informatics and Genomics (BIG) Unit, Laboratory for HealthOmics and Pathway Exploration (HOPE Lab)ChattogramBangladesh
| |
Collapse
|
10
|
Mazeaud C, Pfister S, Owen JE, Pereira HS, Charbonneau F, Robinson ZE, Anton A, Bemis CL, Sow AA, Patel TR, Neufeldt CJ, Scaturro P, Chatel-Chaix L. Zika virus remodels and hijacks IGF2BP2 ribonucleoprotein complex to promote viral replication organelle biogenesis. eLife 2024; 13:RP94347. [PMID: 39565347 DOI: 10.7554/elife.94347] [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] [Indexed: 11/21/2024] Open
Abstract
Zika virus (ZIKV) infection causes significant human disease that, with no approved treatment or vaccine, constitutes a major public health concern. Its life cycle entirely relies on the cytoplasmic fate of the viral RNA genome (vRNA) through a fine-tuned equilibrium between vRNA translation, replication, and packaging into new virions, all within virus-induced replication organelles (vROs). In this study, with an RNA interference (RNAi) mini-screening and subsequent functional characterization, we have identified insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) as a new host dependency factor that regulates vRNA synthesis. In infected cells, IGF2BP2 associates with viral NS5 polymerase and redistributes to the perinuclear viral replication compartment. Combined fluorescence in situ hybridization-based confocal imaging, in vitro binding assays, and immunoprecipitation coupled to RT-qPCR showed that IGF2BP2 directly interacts with ZIKV vRNA 3' nontranslated region. Using ZIKV sub-genomic replicons and a replication-independent vRO induction system, we demonstrated that IGF2BP2 knockdown impairs de novo vRO biogenesis and, consistently, vRNA synthesis. Finally, the analysis of immunopurified IGF2BP2 complex using quantitative mass spectrometry and RT-qPCR revealed that ZIKV infection alters the protein and RNA interactomes of IGF2BP2. Altogether, our data support that ZIKV hijacks and remodels the IGF2BP2 ribonucleoprotein complex to regulate vRO biogenesis and vRNA neosynthesis.
Collapse
Affiliation(s)
- Clément Mazeaud
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada
| | | | - Jonathan E Owen
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, United States
| | - Higor Sette Pereira
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Flavie Charbonneau
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada
| | - Zachary E Robinson
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Anaïs Anton
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada
| | - Cheyanne L Bemis
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, United States
| | - Aïssatou Aïcha Sow
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada
| | - Trushar R Patel
- Department of Chemistry and Biochemistry, Alberta RNA Research and Training Institute, University of Lethbridge, Lethbridge, Canada
| | - Christopher J Neufeldt
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, United States
| | | | - Laurent Chatel-Chaix
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique, Laval, Canada
- Center of Excellence in Research on Orphan Diseases-Fondation Courtois, Quebec, Canada
- Regroupement Intersectoriel de Recherche en Santé de l'Université du Québec, Quebec, Canada
- Swine and Poultry Infectious Diseases Research Centre, Quebec, Canada
| |
Collapse
|
11
|
Carneiro PH, Jimenez-Posada EV, Lopes E, Mohana-Borges R, Biering SB, Harris E. The ApoA1-mimetic peptide 4F blocks flavivirus NS1-triggered endothelial dysfunction and protects against lethal dengue virus challenge. Antiviral Res 2024; 231:106002. [PMID: 39260777 DOI: 10.1016/j.antiviral.2024.106002] [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: 04/26/2024] [Revised: 08/11/2024] [Accepted: 09/07/2024] [Indexed: 09/13/2024]
Abstract
Flavivirus infections result in a variety of outcomes, from clinically inapparent infections to severe, sometimes fatal cases characterized by hemorrhagic manifestations and vascular leakage leading to shock (dengue), meningomyeloencephalitis (West Nile), and congenital abnormalities (Zika). Although there are approved vaccines against several flaviviruses, potentially enhancing cross-reactive immune responses have complicated the development and implementation of vaccines against dengue and Zika viruses, and no specific therapeutics currently exist. The flavivirus nonstructural protein 1 (NS1) is a promising antiviral target because it is a conserved multifunctional virulence factor that directly triggers vascular leak. We previously showed that interactions between NS1 and the ApoA1 lipoprotein modulate DENV infection. Here, we evaluated the potential of the ApoA1-mimetic peptide, 4F, to interfere with endothelial dysfunction mediated by the NS1 protein of dengue, Zika, and West Nile flaviviruses. In an in vitro model consisting of human endothelial cell monolayers, 4F inhibited NS1-induced hyperpermeability, as measured by a transendothelial electrical resistance assay, and prevented NS1-triggered disruption of the endothelial glycocalyx layer. We also demonstrate that treatment with 4F inhibited NS1 interaction with endothelial cells. Finally, we show that 4F protects against lethal DENV challenge in a mouse model, reducing morbidity and mortality in a dose-dependent manner. Our data demonstrate the potential of 4F to inhibit flavivirus NS1-mediated pathology and severe dengue disease in mice and suggest that 4F can also serve as a molecular tool to probe different NS1 functions in vitro and in vivo.
Collapse
Affiliation(s)
- Pedro H Carneiro
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA; Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - E Vanessa Jimenez-Posada
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Eduarda Lopes
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Scott B Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA; Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA.
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA; Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
12
|
Blyden K, Thomas J, Emami-Naeini P, Fashina T, Conrady CD, Albini TA, Carag J, Yeh S. Emerging Infectious Diseases and the Eye: Ophthalmic Manifestations, Pathogenesis, and One Health Perspectives. Int Ophthalmol Clin 2024; 64:39-54. [PMID: 39480207 PMCID: PMC11512616 DOI: 10.1097/iio.0000000000000539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infectious diseases may lead to ocular complications including uveitis, an ocular inflammatory condition with potentially sight-threatening sequelae, and conjunctivitis, inflammation of the conjunctiva. Emerging infectious pathogens with known ocular findings include Ebola virus, Zika virus, Avian influenza virus, Nipah virus, severe acute respiratory syndrome coronaviruses, and Dengue virus. Re-emerging pathogens with ocular findings include Toxoplasma gondii and Plasmodium species that lead to malaria. The concept of One Health involves a collaborative and interdisciplinary approach to achieve optimal health outcomes by combining human, animal, and environmental health factors. This approach examines the interconnected and often complex human-pathogen-intermediate host interactions in infectious diseases that may also result in ocular disease, including uveitis and conjunctivitis. Through a comprehensive review of the literature, we review the ophthalmic findings of emerging infectious diseases, pathogenesis, and One Health perspectives that provide further insight into the disease state. While eye care providers and vision researchers may often focus on key local aspects of disease process and management, additional perspective on host-pathogen-reservoir life cycles and transmission considerations, including environmental factors, may offer greater insight to improve outcomes for affected individuals and stakeholders.
Collapse
Affiliation(s)
- K’Mani Blyden
- Medical College of Georgia, Augusta University, Augusta, GA
| | - Joanne Thomas
- Emory Eye Center, Emory University School of Medicine, Atlanta, GA
- Emory University School of Medicine, Atlanta, GA
| | - Parisa Emami-Naeini
- Department of Ophthalmology, University of California, Davis, Sacramento, CA
| | - Tolulope Fashina
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
| | - Christopher D. Conrady
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE
| | - Thomas A. Albini
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL
| | | | - Steven Yeh
- Department of Ophthalmology, University of Nebraska Medical Center, Omaha, NE
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
13
|
Shanshin DV, Borisevich SS, Shaprova ON, Nesmeyanova VS, Bondar AA, Porozov YB, Khamitov EM, Kolosova EA, Shelemba AA, Ushkalenko ND, Protopopova EV, Sergeev AA, Loktev VB, Shcherbakov DN. Phage Display Revealed the Complex Structure of the Epitope of the Monoclonal Antibody 10H10. Int J Mol Sci 2024; 25:10311. [PMID: 39408641 PMCID: PMC11476565 DOI: 10.3390/ijms251910311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 10/20/2024] Open
Abstract
The annual number of reported human cases of flavivirus infections continues to increase. Measures taken by local healthcare systems and international organizations are not fully successful. In this regard, new approaches to treatment and prevention of flavivirus infections are relevant. One promising approach is to use monoclonal antibody preparations. The mouse mAb 10H10 is capable of interacting with viruses belonging to the genus Orthoflavivirus which are pathogenic to humans. ELISA and molecular modeling data can indicate that mAb 10H10 recognizes the fusion loop region of E protein. The KD of interaction between the mAb 10H10 and recombinant analogs of the E protein of the tick-borne encephalitis (TBEV), Zika (ZIKV) and dengue (DENV) viruses range from 1.5 to 4 nM. The aim of this study was to map the epitope of this antibody using phage display technology. After three rounds of biopanning, 60 individual phage clones were chosen. The amino acid sequences of the selected peptides were conveniently divided into five groups. Based on the selected peptides, bacteriophages were obtained carrying peptides on the surfaces of the pIII and pVIII proteins, which were tested for binding to the antibody in ELISA. Thus, the epitope of the mAb 10H10 is the highly conserved region 98-DRGWGNXXGLFGK-110 of the flavivirus E protein. The structures of the complexes of the identified peptides with the antibody paratope are proposed using the molecular docking and dynamics methods.
Collapse
Affiliation(s)
- Daniil V. Shanshin
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Sophia S. Borisevich
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
- Synchrotron Radiation Facility—Siberian Circular Photon Source “SKlF” Boreskov Institute of Catalysis of Siberian Branch of the Russian Academy of Sciences, Koltsovo 630559, Russia
| | - Olga N. Shaprova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Valentina S. Nesmeyanova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Alexander A. Bondar
- Genomics Core Facility, Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk 630090, Russia;
| | - Yuri B. Porozov
- Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street, Moscow 125315, Russia;
- Advitam Laboratory, 11108 Belgrade, Serbia
| | - Edward M. Khamitov
- Laboratory of Physical and Chemical Methods of Analysis, Ufa Institute of Chemistry UFRS RAS, Ufa 450054, Russia;
| | - Evgeniia A. Kolosova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| | - Arseniya A. Shelemba
- Federal State Budgetary Scientific Institution “Federal Research Center for Fundamental and Translational Medicine”, Novosibirsk 630117, Russia;
| | - Nikita D. Ushkalenko
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Elena V. Protopopova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Artemiy A. Sergeev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Valery B. Loktev
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
| | - Dmitriy N. Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo 630559, Russia; (D.N.S.); (O.N.S.); (V.S.N.); (E.A.K.); (N.D.U.); (E.V.P.); (A.A.S.); (V.B.L.)
- Research Institute of Biological Medicine Center for Recombinant Technologies, Altay State University, Barnaul 656049, Russia
| |
Collapse
|
14
|
Altayb HN, Alatawi HA. Employing Machine Learning-Based QSAR for Targeting Zika Virus NS3 Protease: Molecular Insights and Inhibitor Discovery. Pharmaceuticals (Basel) 2024; 17:1067. [PMID: 39204173 PMCID: PMC11359100 DOI: 10.3390/ph17081067] [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: 07/15/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
Zika virus infection is a mosquito-borne viral disease that has become a global health concern recently. Zika virus belongs to the Flavivirus genus and is primarily transmitted by Aedes mosquitoes. Prevention of Zika virus infection involves avoiding mosquito bites by using repellent, wearing protective clothing, and staying in screened areas, especially for pregnant women. Treatment focuses on managing symptoms with rest, fluids, and acetaminophen, with close monitoring for pregnant women. Currently, there is no specific antiviral treatment or vaccine for the Zika virus, highlighting the importance of prevention strategies to control its spread. Therefore, in this study, the Zika virus non-structural protein NS3 was targeted to inhibit Zika infection by identifying the novel inhibitor through an in silico approach. Here, 2864 natural compounds were screened using a machine learning-based QSAR model, and later docking was performed to select the potential target. Subsequently, Tanimoto similarity and clustering were performed to obtain the potential target. The three most potential compounds were obtained: (a) 5297, (b) 432449, and (c) 85137543. The protein-ligand complex's stability and flexibility were then investigated by dynamic modelling. The 300 ns simulation showed that 5297 exhibited the steadiest deviation and constant creation of hydrogen bonds. Compared to the other compounds, 5297 demonstrated a superior binding free energy (ΔG = -20.81 kcal/mol) with the protein when the MM/GBSA technique was used. The study determined that 5297 showed significant therapeutic potential and justifies further experimental investigation as a possible inhibitor of the NS2B-NS3 protease target implicated in Zika virus infection.
Collapse
Affiliation(s)
- Hisham N. Altayb
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Hanan Ali Alatawi
- Department of Biological Sciences, University Collage of Haqel, University of Tabuk, Tabuk 71491, Saudi Arabia;
| |
Collapse
|
15
|
Adalat S, Hajat S, Ikiz B. Climate change has serious implications for children's brain health. BMJ 2024; 386:q1588. [PMID: 39038838 DOI: 10.1136/bmj.q1588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Affiliation(s)
| | - Shakoor Hajat
- Centre on Climate Change and Planetary Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Burcin Ikiz
- Neuro Climate Working Group, Global Consortium on Climate and Health Education, Columbia University, New York, USA
| |
Collapse
|
16
|
Nogueira CDO, Lopes da Silva MO, de Lima EV, Christoff RR, Gavino-Leopoldino D, Lemos FS, da Silva NE, Da Poian AT, Assunção-Miranda I, Figueiredo CP, Clarke JR. Immunosuppression-induced Zika virus reactivation causes brain inflammation and behavioral deficits in mice. iScience 2024; 27:110178. [PMID: 38993676 PMCID: PMC11237861 DOI: 10.1016/j.isci.2024.110178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/27/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Zika virus (ZIKV) is a neurotropic flavivirus that can persist in several tissues. The late consequences of ZIKV persistence and whether new rounds of active replication can occur, remain unaddressed. Here, we investigated whether neonatally ZIKV-infected mice are susceptible to viral reactivation in adulthood. We found that when ZIKV-infected mice are treated with immunosuppressant drugs, they present increased susceptibility to chemically induced seizures. Levels of subgenomic flavivirus RNAs (sfRNAs) were increased, relative to the amounts of genomic RNAs, in the brains of mice following immunosuppression and were associated with changes in cytokine expression. We investigated the impact of immunosuppression on the testicles and found that ZIKV genomic RNA levels are increased in mice following immunosuppression, which also caused significant testicular damage. These findings suggest that ZIKV can establish new rounds of active replication long after acute stages of disease, so exposed patients should be monitored to ensure complete viral eradication.
Collapse
Affiliation(s)
- Clara de O Nogueira
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | | | - Emanuelle V de Lima
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Raíssa Rilo Christoff
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Daniel Gavino-Leopoldino
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Felipe S Lemos
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Nicolas E da Silva
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Iranaia Assunção-Miranda
- Instituto de Microbiologia Paulo de Goes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Claudia P Figueiredo
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| | - Julia R Clarke
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941902, RJ, Brazil
| |
Collapse
|
17
|
Miranda-López A, González-Ortega O, Govea-Alonso DO, Betancourt-Mendiola L, Comas-García M, Rosales-Mendoza S. Rational design and production of a chimeric antigen targeting Zika virus that induces neutralizing antibodies in mice. Vaccine 2024; 42:3674-3683. [PMID: 38749821 DOI: 10.1016/j.vaccine.2024.04.080] [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: 12/14/2023] [Revised: 03/14/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024]
Abstract
The Zika virus (ZIKV) is considered a public health problem worldwide due to its association with the development of microcephaly and the Guillain-Barré syndrome. Currently, there is no specific treatment or vaccine approved to combat this disease, and thus, developing safe and effective vaccines is a relevant goal. In this study, a multi-epitope protein called rpZDIII was designed based on a series of ZIKV antigenic sequences, a bacterial carrier, and linkers. The analysis of the predicted 3D structure of the rpZDIII chimeric antigen was performed on the AlphaFold 2 server, and it was produced in E. coli and purified from inclusion bodies, followed by solubilization and refolding processes. The yield achieved for rpZDIII was 11 mg/L in terms of pure soluble recombinant protein per liter of fermentation. rpZDIII was deemed immunogenic since it induced serum IgG and IgM responses in mice upon subcutaneous immunization in a three-dose scheme. Moreover, sera from mice immunized with rpZDIII showed neutralizing activity against ZIKV. Therefore, this study reveals rpZDIII as a promising immunogen for the development of a rationally designed multi-epitope vaccine against ZIKV, and completion of its preclinical evaluation is guaranteed.
Collapse
Affiliation(s)
- Arleth Miranda-López
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México
| | - Omar González-Ortega
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Dania O Govea-Alonso
- Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Lourdes Betancourt-Mendiola
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México
| | - Mauricio Comas-García
- Sección de Microscopía de Alta Resolución, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210, México; Facultad de Ciencias, Universidad Autónoma de San Luis Potosí, Av. Parque Chapultepec 1570, San Luis, S.L.P., San Luis Potosí 78210, México.
| | - Sergio Rosales-Mendoza
- Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP 78210, México; Sección de Biotecnología, Centro de Investigación en Ciencias de la Salud y Biomedicina, Universidad Autónoma de San Luis Potosí, Av. Sierra Leona 550, Lomas 2ª. Sección, San Luis Potosí 78210 México.
| |
Collapse
|
18
|
Sapkal G, Deshpande GR, Gupta N, Deshpande K, Sharma S, Tandale B, Srivastava R, Vidhate S, Khutwad K, Tilekar BN. Harmonization of Zika serological assays and comparative evaluation of two commercial ZIKA IgG ELISA kits. Diagn Microbiol Infect Dis 2024; 109:116238. [PMID: 38554539 DOI: 10.1016/j.diagmicrobio.2024.116238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/27/2024] [Accepted: 02/27/2024] [Indexed: 04/01/2024]
Abstract
The interpretation for Zika virus serology results is challenging due to high antibody cross reactivity with other flaviviruses. This limits availability of reliable and accurate methods for serosurveillance studies to understand the disease burden. Therefore, we conducted study to harmonize anti-Zika IgG antibody detection assays with 1st WHO International Standard (16/352) and working standard (16/320) for anti-Zika virus antibody.Additionally, evaluated NuGenTMZIKA-IgG and NovaLisa®ZIKA virus IgG-Capture ELISA using a panel of 278 seraFurther, 106 samples positive for other-flavi viruses were taken for assessing cross-reactivity of the assay, all serums were further tested by Zika-PRNT. The results of this study indicates satisfactory performance of both the assays. Serological and neutralization assays were calibrated according to the international standards. This will help in understanding antibody dynamics in serosurveillance and vaccine studies. However the performance of the kits with possibilities of cross-reactivity will have to be verified by coupling ZIKV and DENV specific ELISA.
Collapse
Affiliation(s)
- Gajanan Sapkal
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India.
| | | | - Nivedita Gupta
- Indian Council of Medical Research, New Delhi 110001,India
| | - Ketki Deshpande
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - Sharada Sharma
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - Babasaheb Tandale
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - Rashi Srivastava
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - Shankar Vidhate
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - Kirtee Khutwad
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| | - B N Tilekar
- ICMR-National Institute of Virology, Pune 411021, Maharashtra, India
| |
Collapse
|
19
|
Wongsawat J, Thamthitiwat S, Hicks VJ, Uttayamakul S, Teepruksa P, Sawatwong P, Skaggs B, Mock PA, MacArthur JR, Suya I, Sapchookul P, Kitsutani P, Lo TQ, Vachiraphan A, Kovavisarach E, Rhee C, Darun P, Saepueng K, Waisaen C, Jampan D, Sriboonrat P, Palanuwong B, Sukbut P, Areechokchai D, Pittayawonganon C, Iamsirithaworn S, Bloss E, Rao CY. Characteristics, risk factors, and outcomes related to Zika virus infection during pregnancy in Northeastern Thailand: A prospective pregnancy cohort study, 2018-2020. PLoS Negl Trop Dis 2024; 18:e0012176. [PMID: 38758964 PMCID: PMC11139345 DOI: 10.1371/journal.pntd.0012176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 05/30/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND In response to the 2015-2016 Zika virus (ZIKV) outbreak and the causal relationship established between maternal ZIKV infection and adverse infant outcomes, we conducted a cohort study to estimate the incidence of ZIKV infection in pregnancy and assess its impacts in women and infants. METHODOLOGY/PRINCIPAL FINDINGS From May 2018-January 2020, we prospectively followed pregnant women recruited from 134 participating hospitals in two non-adjacent provinces in northeastern Thailand. We collected demographic, clinical, and epidemiologic data and blood and urine at routine antenatal care visits until delivery. ZIKV infections were confirmed by real-time reverse transcriptase polymerase chain reaction (rRT-PCR). Specimens with confirmed ZIKV underwent whole genome sequencing. Among 3,312 women enrolled, 12 (0.36%) had ZIKV infections, of which two (17%) were detected at enrollment. Ten (83%, 3 in 2nd and 7 in 3rd trimester) ZIKV infections were detected during study follow-up, resulting in an infection rate of 0.15 per 1,000 person-weeks (95% CI: 0.07-0.28). The majority (11/12, 91.7%) of infections occurred in one province. Persistent ZIKV viremia (42 days) was found in only one woman. Six women with confirmed ZIKV infections were asymptomatic until delivery. Sequencing of 8 ZIKV isolates revealed all were of Asian lineage. All 12 ZIKV infected women gave birth to live, full-term infants; the only observed adverse birth outcome was low birth weight in one (8%) infant. Pregnancies in 3,300 ZIKV-rRT-PCR-negative women were complicated by 101 (3%) fetal deaths, of which 67 (66%) had miscarriages and 34 (34%) had stillbirths. There were no differences between adverse fetal or birth outcomes of live infants born to ZIKV-rRT-PCR-positive mothers compared to live infants born to ZIKV-rRT-PCR-negative mothers. CONCLUSIONS/SIGNIFICANCE Confirmed ZIKV infections occurred infrequently in this large pregnancy cohort and observed adverse maternal and birth outcomes did not differ between mothers with and without confirmed infections.
Collapse
Affiliation(s)
- Jurai Wongsawat
- Thailand Ministry of Public Health, Department of Disease Control, Nonthaburi, Thailand
| | - Somsak Thamthitiwat
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Victoria J. Hicks
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Sumonmal Uttayamakul
- Thailand Ministry of Public Health, Department of Disease Control, Nonthaburi, Thailand
| | - Phanthaneeya Teepruksa
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Pongpun Sawatwong
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Beth Skaggs
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Philip A. Mock
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - John R. MacArthur
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Inthira Suya
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Patranuch Sapchookul
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Paul Kitsutani
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Terrence Q. Lo
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Apichart Vachiraphan
- Thailand Ministry of Public Health, Department of Disease Control, Nonthaburi, Thailand
| | - Ekachai Kovavisarach
- Thailand Ministry of Public Health, Department of Medical Services, Nonthaburi, Thailand
| | - Chulwoo Rhee
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Pamorn Darun
- Bueng Kan Provincial Public Health Office, Bueng Kan, Thailand
| | | | - Chamnan Waisaen
- Bueng Kan Provincial Public Health Office, Bueng Kan, Thailand
| | | | | | | | | | - Darin Areechokchai
- Thailand Ministry of Public Health, Department of Disease Control, Nonthaburi, Thailand
| | | | - Sopon Iamsirithaworn
- Thailand Ministry of Public Health, Department of Disease Control, Nonthaburi, Thailand
| | - Emily Bloss
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| | - Carol Y. Rao
- Thailand Ministry of Public Health–US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- US Centers for Disease Control and Prevention, Division of Global Health Protection, Atlanta, Georgia, United States of America
| |
Collapse
|
20
|
Chen X, Li RT, Chen RY, Shi PD, Liu ZX, Lou YN, Wu M, Zhang RR, Tang W, Li XF, Qin CF. The subgenomic flaviviral RNA suppresses RNA interference through competing with siRNAs for binding RISC components. J Virol 2024; 98:e0195423. [PMID: 38289102 PMCID: PMC10878275 DOI: 10.1128/jvi.01954-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 02/21/2024] Open
Abstract
During the life cycle of mosquito-borne flaviviruses, substantial subgenomic flaviviral RNA (sfRNA) is produced via incomplete degradation of viral genomic RNA by host XRN1. Zika virus (ZIKV) sfRNA has been detected in mosquito and mammalian somatic cells. Human neural progenitor cells (hNPCs) in the developing brain are the major target cells of ZIKV, and antiviral RNA interference (RNAi) plays a critical role in hNPCs. However, whether ZIKV sfRNA was produced in ZIKV-infected hNPCs as well as its function remains not known. In this study, we demonstrate that abundant sfRNA was produced in ZIKV-infected hNPCs. RNA pulldown and mass spectrum assays showed ZIKV sfRNA interacted with host proteins RHA and PACT, both of which are RNA-induced silencing complex (RISC) components. Functionally, ZIKV sfRNA can antagonize RNAi by outcompeting small interfering RNAs (siRNAs) in binding to RHA and PACT. Furthermore, the 3' stem loop (3'SL) of sfRNA was responsible for RISC components binding and RNAi inhibition, and 3'SL can enhance the replication of a viral suppressor of RNAi (VSR)-deficient virus in a RHA- and PACT-dependent manner. More importantly, the ability of binding to RISC components is conversed among multiple flaviviral 3'SLs. Together, our results identified flavivirus 3'SL as a potent VSR in RNA format, highlighting the complexity in virus-host interaction during flavivirus infection.IMPORTANCEZika virus (ZIKV) infection mainly targets human neural progenitor cells (hNPCs) and induces cell death and dysregulated cell-cycle progression, leading to microcephaly and other central nervous system abnormalities. RNA interference (RNAi) plays critical roles during ZIKV infections in hNPCs, and ZIKV has evolved to encode specific viral proteins to antagonize RNAi. Herein, we first show that abundant sfRNA was produced in ZIKV-infected hNPCs in a similar pattern to that in other cells. Importantly, ZIKV sfRNA acts as a potent viral suppressor of RNAi (VSR) by competing with siRNAs for binding RISC components, RHA and PACT. The 3'SL of sfRNA is responsible for binding RISC components, which is a conserved feature among mosquito-borne flaviviruses. As most known VSRs are viral proteins, our findings highlight the importance of viral non-coding RNAs during the antagonism of host RNAi-based antiviral innate immunity.
Collapse
Affiliation(s)
- Xiang Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Rui-Ting Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ru-Yi Chen
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Pan-Deng Shi
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Zi-Xin Liu
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Ya-Nan Lou
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Mei Wu
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Rong-Rong Zhang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Wei Tang
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiao-Feng Li
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
- Research Unit of Discovery and Tracing of Natural Focus Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
21
|
Kim IJ, Gonzalez O, Tighe MP, Lanthier PA, Clark MJ, Travis KL, Low-Beer TC, Lanzer KG, Bernacki DT, Szaba FM, De La Barrera RA, Dussupt V, Mendez-Rivera L, Krebs SJ, Ross CN, Mdaki SD, Brasky KM, Layne-Colon D, Tardif SD, Thomas SJ, Modjarrad K, Blackman MA, Patterson JL. Protective efficacy of a Zika purified inactivated virus vaccine candidate during pregnancy in marmosets. NPJ Vaccines 2024; 9:35. [PMID: 38368443 PMCID: PMC10874403 DOI: 10.1038/s41541-024-00824-0] [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: 07/10/2023] [Accepted: 01/30/2024] [Indexed: 02/19/2024] Open
Abstract
Zika virus (ZIKV) infection during pregnancy poses significant threats to maternal and fetal health, leading to intrauterine fetal demise and severe developmental malformations that constitute congenital Zika syndrome (CZS). As such, the development of a safe and effective ZIKV vaccine is a critical public health priority. However, the safety and efficacy of such a vaccine during pregnancy remain uncertain. Historically, the conduct of clinical trials in pregnant women has been challenging. Therefore, clinically relevant animal pregnancy models are in high demand for testing vaccine efficacy. We previously reported that a marmoset pregnancy model of ZIKV infection consistently demonstrated vertical transmission from mother to fetus during pregnancy. Using this marmoset model, we also showed that vertical transmission could be prevented by pre-pregnancy vaccination with Zika purified inactivated virus (ZPIV) vaccine. Here, we further examined the efficacy of ZPIV vaccination during pregnancy. Vaccination during pregnancy elicited virus neutralizing antibody responses that were comparable to those elicited by pre-pregnancy vaccination. Vaccination also reduced placental pathology, viral burden and vertical transmission of ZIKV during pregnancy, without causing adverse effects. These results provide key insights into the safety and efficacy of ZPIV vaccination during pregnancy and demonstrate positive effects of vaccination on the reduction of ZIKV infection, an important advance in preparedness for future ZIKV outbreaks.
Collapse
Affiliation(s)
- In-Jeong Kim
- Trudeau Institute, Inc., Saranac Lake, NY, 12983, USA.
| | - Olga Gonzalez
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | | | | | | | | | | | | | | | - Frank M Szaba
- Trudeau Institute, Inc., Saranac Lake, NY, 12983, USA
| | - Rafael A De La Barrera
- Pilot Bioproduction Facility, Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Vincent Dussupt
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Letzibeth Mendez-Rivera
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Shelly J Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Corinna N Ross
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Stephanie D Mdaki
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
- Science and Technology, Joint Base San Antonio-Fort Sam AFB, San Antonio, TX, 78236, USA
| | - Kathleen M Brasky
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Donna Layne-Colon
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Suzette D Tardif
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Stephen J Thomas
- Institute for Global Health and Translational Sciences, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Pfizer Inc. Vaccine Research and Development, Pearl River, NY, 10965, USA
| | | | - Jean L Patterson
- Southwest National Primate Center, Texas Biomedical Research Institute, San Antonio, TX, 78227, USA.
| |
Collapse
|
22
|
Kim IJ, Tighe MP, Lanthier PA, Clark MJ, De La Barrera RA, Dussupt V, Mendez-Rivera L, Krebs SJ, Travis KL, Low-Beer TC, Cookenham TS, Lanzer KG, Bernacki DT, Szaba FM, Schneck AA, Ward J, Thomas SJ, Modjarrad K, Blackman MA. Zika purified inactivated virus (ZPIV) vaccine reduced vertical transmission in pregnant immunocompetent mice. NPJ Vaccines 2024; 9:32. [PMID: 38360793 PMCID: PMC10869681 DOI: 10.1038/s41541-024-00823-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/30/2024] [Indexed: 02/17/2024] Open
Abstract
Zika virus (ZIKV) is a significant threat to pregnant women and their fetuses as it can cause severe birth defects and congenital neurodevelopmental disorders, referred to as congenital Zika syndrome (CZS). Thus, a safe and effective ZIKV vaccine for pregnant women to prevent in utero ZIKV infection is of utmost importance. Murine models of ZIKV infection are limited by the fact that immunocompetent mice are resistant to ZIKV infection. As such, interferon-deficient mice have been used in some preclinical studies to test the efficacy of ZIKV vaccine candidates against lethal virus challenge. However, interferon-deficient mouse models have limitations in assessing the immunogenicity of vaccines, necessitating the use of immunocompetent mouse pregnancy models. Using the human stat2 knock-in (hSTAT2KI) mouse pregnancy model, we show that vaccination with a purified formalin-inactivated Zika virus (ZPIV) vaccine prior to pregnancy successfully prevented vertical transmission. In addition, maternal immunity protected offspring against postnatal challenge for up to 28 days. Furthermore, passive transfer of human IgG purified from hyper-immune sera of ZPIV vaccinees prevented maternal and fetal ZIKV infection, providing strong evidence that the neutralizing antibody response may serve as a meaningful correlate of protection.
Collapse
Affiliation(s)
- In-Jeong Kim
- Trudeau Institute, Inc., Saranac Lake, NY, 12983, USA.
| | | | | | | | - Rafael A De La Barrera
- Pilot Bioproduction Facility, Center for Enabling Capabilities, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Vincent Dussupt
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Letzibeth Mendez-Rivera
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | - Shelly J Krebs
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- U.S. Military HIV Research Program, Center of Infectious Disease Research, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, 20817, USA
| | | | | | | | | | | | - Frank M Szaba
- Trudeau Institute, Inc., Saranac Lake, NY, 12983, USA
| | | | - Jerrold Ward
- Global VetPathology, Montgomery Village, MD, 20886, USA
| | - Stephen J Thomas
- Institute for Global Health and Translational Sciences, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| | - Kayvon Modjarrad
- Emerging Infectious Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Pfizer Inc. Vaccine Research and Development, Pearl River, NY, 10965, USA
| | | |
Collapse
|
23
|
Ahmed S, Sultana S, Kundu S, Alam SS, Hossan T, Islam MA. Global Prevalence of Zika and Chikungunya Coinfection: A Systematic Review and Meta-Analysis. Diseases 2024; 12:31. [PMID: 38391778 PMCID: PMC10888207 DOI: 10.3390/diseases12020031] [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: 11/16/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Zika virus (ZIKV) and chikungunya virus (CHIKV) are arthropod-borne viruses with significant pathogenicity, posing a substantial health and economic burden on a global scale. Moreover, ZIKV-CHIKV coinfection imposes additional therapeutic challenges as there is no specific treatment for ZIKV or CHIKV infection. While a growing number of studies have documented the ZIKV-CHIKV coinfection, there is currently a lack of conclusive reports on this coinfection. Therefore, we performed a systematic review and meta-analysis to determine the true statistics of ZIKV-CHIKV coinfection in the global human population. Relevant studies were searched for in PubMed, Scopus, and Google Scholar without limitation in terms of language or publication date. A total of 33 studies containing 41,460 participants were included in this meta-analysis. The study protocol was registered with PROSPERO under the registration number CRD42020176409. The pooled prevalence and confidence intervals of ZIKV-CHIKV coinfection were computed using a random-effects model. The study estimated a combined global prevalence rate of 1.0% [95% CI: 0.7-1.2] for the occurrence of ZIKV-CHIKV coinfection. The region of North America (Mexico, Haiti, and Nicaragua) and the country of Haiti demonstrated maximum prevalence rates of 2.8% [95% CI: 1.5-4.1] and 3.5% [95% CI: 0.2-6.8], respectively. Moreover, the prevalence of coinfection was found to be higher in the paediatric group (2.1% [95% CI: 0.0-4.2]) in comparison with the adult group (0.7% [95% CI: 0.2-1.1]). These findings suggest that the occurrence of ZIKV-CHIKV coinfection varies geographically and by age group. The results of this meta-analysis will guide future investigations seeking to understand the underlying reasons for these variations and the causes of coinfection and to develop targeted prevention and control strategies.
Collapse
Affiliation(s)
- Saleh Ahmed
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Shabiha Sultana
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Shoumik Kundu
- Department of Chemistry and Biochemistry, Texas Tech University, 2500 Broadway St., Lubbock, TX 79409, USA
| | - Sayeda Sadia Alam
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
| | - Tareq Hossan
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka 1342, Bangladesh
- Department of Internal Medicine, Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Md Asiful Islam
- WHO Collaborating Centre for Global Women's Health, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
24
|
Alger J, Cafferata ML, López R, Wiggins LD, Callejas A, Castillo M, Fúnes J, Rico F, Valencia D, Varela D, Alvarez Z, Berrueta M, Bock H, Bustillo C, Calderón A, Ciganda A, García-Aguilar J, García K, Gibbons L, Gilboa SM, Harville EW, Hernández G, López W, Lorenzana I, Luque MT, Maldonado C, Moore C, Ochoa C, Parham L, Pastrana K, Paternina-Caicedo A, Rodríguez H, Stella C, Tannis AF, Wesson DM, Zúniga C, Tong VT, Buekens P. Neurodevelopmental assessment of normocephalic children born to Zika virus exposed and unexposed pregnant people. Pediatr Res 2024; 95:566-572. [PMID: 38057577 PMCID: PMC11045253 DOI: 10.1038/s41390-023-02951-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Studies examining the association between in utero Zika virus (ZIKV) exposure and child neurodevelopmental outcomes have produced varied results. METHODS We aimed to assess neurodevelopmental outcomes among normocephalic children born from pregnant people enrolled in the Zika in Pregnancy in Honduras (ZIPH) cohort study, July-December 2016. Enrollment occurred during the first prenatal visit. Exposure was defined as prenatal ZIKV IgM and/or ZIKV RNA result at enrollment. Normocephalic children, >6 months old, were selected for longitudinal follow-up using the Bayley Scales of Infant and Toddler Development (BSID-III) and the Ages & Stages Questionnaires: Social-Emotional (ASQ:SE-2). RESULTS One hundred fifty-two children were assessed; after exclusion, 60 were exposed and 72 were unexposed to ZIKV during pregnancy. Twenty children in the exposed group and 21 children in the unexposed group had a composite score <85 in any of the BSID-III domains. Although exposed children had lower cognitive and language scores, differences were not statistically significant. For ASQ:SE-2 assessment, there were not statistically significant differences between groups. CONCLUSIONS This study found no statistically significant differences in the neurodevelopment of normocephalic children between in utero ZIKV exposed and unexposed. Nevertheless, long-term monitoring of children with in utero ZIKV exposure is warranted. IMPACT This study found no statistically significant differences in the neurodevelopment in normocephalic children with in utero Zika virus exposure compared to unexposed children, although the exposed group showed lower cognitive and language scores that persisted after adjustment by maternal age and education and after excluding children born preterm and low birth weight from the analysis. Children with prenatal Zika virus exposure, including those normocephalic and have no evidence of abnormalities at birth, should be monitored for neurodevelopmental delays. Follow-up is important to be able to detect developmental abnormalities that might not be detected earlier in life.
Collapse
Affiliation(s)
- Jackeline Alger
- Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras.
| | | | - Raquel López
- Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | - Lisa D Wiggins
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Allison Callejas
- Servicio de Neonatología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Mario Castillo
- Servicio de Neonatología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Jenny Fúnes
- Servicio de Neonatología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Fátima Rico
- Departamento de Pediatría, Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Diana Valencia
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Douglas Varela
- Servicio de Neurología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Zulma Alvarez
- Unidad de Vigilancia de la Salud, Región Sanitaria Metropolitana del Distrito Central, Secretaría de Salud de Honduras, Tegucigalpa, Honduras
| | - Mabel Berrueta
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Harry Bock
- Dirección General, Región Sanitaria Metropolitana del Distrito Central, currently Centro de Salud Dra. Nerza Paz, Región Sanitaria Metropolitana del Distrito Central, Secretaría de Salud de Honduras, Tegucigalpa, Honduras
| | - Carolina Bustillo
- Departamento de Ginecología y Obstetricia, Hospital Escuela, Tegucigalpa, Honduras
| | - Alejandra Calderón
- Centro de Salud Alonso Suazo, Región Sanitaria Metropolitana del Distrito Central, currently Centro de Salud Villanueva, Región Sanitaria Metropolitana del Distrito Central, Secretaría de Salud de Honduras, Tegucigalpa, Honduras
| | - Alvaro Ciganda
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Jorge García-Aguilar
- Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | - Kimberly García
- Centro de Investigaciones Genéticas, Instituto de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Luz Gibbons
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | - Suzanne M Gilboa
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Emily W Harville
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Gustavo Hernández
- Departamento de Pediatría, Hospital de Especialidades San Felipe, Tegucigalpa, Honduras
| | - Wendy López
- Instituto de Enfermedades Infecciosas y Parasitología Antonio Vidal, Tegucigalpa, Honduras
| | - Ivette Lorenzana
- Centro de Investigaciones Genéticas, Instituto de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Marco T Luque
- Servicio de Infectología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Carlos Maldonado
- Servicio de Oftalmología, Departamento de Pediatría, Hospital Escuela, Tegucigalpa, Honduras
| | - Cynthia Moore
- Goldbelt Professional Services, LLC, Chesapeake, VA, USA
| | - Carlos Ochoa
- Servicio de Maternidad, Hospital de Especialidades San Felipe, Tegucigalpa, Honduras
| | - Leda Parham
- Centro de Investigaciones Genéticas, Instituto de Investigaciones en Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Karla Pastrana
- Departamento de Ginecología y Obstetricia, Hospital Escuela, Tegucigalpa, Honduras
| | - Angel Paternina-Caicedo
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Heriberto Rodríguez
- Departamento de Ginecología y Obstetricia, Hospital Escuela, Tegucigalpa, Honduras
| | - Candela Stella
- Instituto de Efectividad Clínica y Sanitaria, Buenos Aires, Argentina
| | | | - Dawn M Wesson
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Concepción Zúniga
- Departamento de Vigilancia de la Salud, Hospital Escuela, Tegucigalpa, Honduras
| | - Van T Tong
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Pierre Buekens
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| |
Collapse
|
25
|
STOKES CALEB, J. MELVIN ANN. Viral Infections of the Fetus and Newborn. AVERY'S DISEASES OF THE NEWBORN 2024:450-486.e24. [DOI: 10.1016/b978-0-323-82823-9.00034-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
26
|
Hu D, Tian Y, Xu J, Xie D, Wang Y, Liu M, Wang Y, Yang L. Oncolytic viral therapy as promising immunotherapy against glioma. MEDCOMM – FUTURE MEDICINE 2023; 2. [DOI: 10.1002/mef2.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/22/2023] [Indexed: 03/19/2025]
Abstract
AbstractGlioma is a common primary central nervous system malignant tumor in clinical, traditional methods such as surgery and chemoradiotherapy are not effective in treatment. Therefore, more effective treatments need to be found. Oncolytic viruses (OVs) are a new type of immunotherapy that selectively infects and kills tumor cells instead of normal cells. OVs can mediate antitumor immune responses through a variety of mechanisms, and have the ability to activate antitumor immune responses, transform the tumor microenvironment from “cold” to “hot,” and enhance the efficacy of immune checkpoint inhibitors. Recently, a large number of preclinical and clinical studies have shown that OVs show great prospects in the treatment of gliomas. In this review, we summarize the current status of glioma therapies with a focus on OVs. First, this article introduces the current status of treatment of glioma and their respective shortcomings. Then, the important progress of OVs of in clinical trials of glioma is summarized. Finally, the urgent challenges of oncolytic virus treatment for glioma are sorted out, and related solutions are proposed. This review will help to further promote the use of OVs in the treatment of glioma.
Collapse
Affiliation(s)
- Die Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yaomei Tian
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- College of Bioengineering Sichuan University of Science & Engineering Zigong China
| | - Jie Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Daoyuan Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yusi Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Mohan Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Yuanda Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| |
Collapse
|
27
|
Moadab G, Pittet F, Bennett JL, Taylor CL, Fiske O, Singapuri A, Coffey LL, Van Rompay KKA, Bliss-Moreau E. Prenatal Zika virus infection has sex-specific effects on infant physical development and mother-infant social interactions. Sci Transl Med 2023; 15:eadh0043. [PMID: 37878673 DOI: 10.1126/scitranslmed.adh0043] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023]
Abstract
There is enormous variation in the extent to which fetal Zika virus (fZIKV) infection affects the developing brain. Despite the neural consequences of fZIKV infection observed in people and animal models, many open questions about the relationship between infection dynamics and fetal and infant development remain. To further understand how ZIKV affects the developing nervous system and the behavioral consequences of prenatal infection, we adopted a nonhuman primate model of fZIKV infection in which we inoculated pregnant rhesus macaques and their fetuses with ZIKV in the early second trimester of fetal development. We then tracked their health across gestation and characterized infant development across the first month of life. ZIKV-infected pregnant mothers had long periods of viremia and mild changes to their hematological profiles. ZIKV RNA concentrations, an indicator of infection magnitude, were higher in mothers whose fetuses were male, and the magnitude of ZIKV RNA in the mothers' plasma or amniotic fluid predicted infant outcomes. The magnitude of ZIKV RNA was negatively associated with infant growth across the first month of life, affecting males' growth more than females' growth, although for most metrics, both males and females evidenced slower growth rates as compared with control animals whose mothers were not ZIKV inoculated. Compared with control infants, fZIKV infants also spent more time with their mothers during the first month of life, a social behavior difference that may have long-lasting consequences on psychosocial development during childhood.
Collapse
Affiliation(s)
- Gilda Moadab
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Florent Pittet
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Jeffrey L Bennett
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Christopher L Taylor
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Olivia Fiske
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| | - Anil Singapuri
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Lark L Coffey
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
- Department of Pathology, Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
| | - Eliza Bliss-Moreau
- Department of Psychology, University of California, Davis, Davis, CA 95616, USA
- California National Primate Research Center, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
28
|
Kulkarni A, Delgadillo FM, Gayathrinathan S, Grajeda BI, Roy S. Current Status of Omics Studies Elucidating the Features of Reproductive Biology in Blood-Feeding Insects. INSECTS 2023; 14:802. [PMID: 37887814 PMCID: PMC10607566 DOI: 10.3390/insects14100802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
Female insects belonging to the genera Anopheles, Aedes, Glossina, and Rhodnius account for the majority of global vector-borne disease mortality. In response to mating, these female insects undergo several molecular, physiological, and behavioral changes. Studying the dynamic post-mating molecular responses in these insects that transmit human diseases can lead to the identification of potential targets for the development of novel vector control methods. With the continued advancements in bioinformatics tools, we now have the capability to delve into various physiological processes in these insects. Here, we discuss the availability of multiple datasets describing the reproductive physiology of the common blood-feeding insects at the molecular level. Additionally, we compare the male-derived triggers transferred during mating to females, examining both shared and species-specific factors. These triggers initiate post-mating genetic responses in female vectors, affecting not only their reproductive success but also disease transmission.
Collapse
Affiliation(s)
- Aditi Kulkarni
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Frida M. Delgadillo
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Environmental Science and Engineering Ph.D. Program, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Sharan Gayathrinathan
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Brian I. Grajeda
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Biosciences Ph.D. Program, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Sourav Roy
- Department of Biological Sciences, University of Texas at El Paso, El Paso, TX 79968, USA; (A.K.); (F.M.D.); (S.G.); (B.I.G.)
- Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| |
Collapse
|
29
|
Diani E, Lagni A, Lotti V, Tonon E, Cecchetto R, Gibellini D. Vector-Transmitted Flaviviruses: An Antiviral Molecules Overview. Microorganisms 2023; 11:2427. [PMID: 37894085 PMCID: PMC10608811 DOI: 10.3390/microorganisms11102427] [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: 08/17/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Flaviviruses cause numerous pathologies in humans across a broad clinical spectrum with potentially severe clinical manifestations, including hemorrhagic and neurological disorders. Among human flaviviruses, some viral proteins show high conservation and are good candidates as targets for drug design. From an epidemiological point of view, flaviviruses cause more than 400 million cases of infection worldwide each year. In particular, the Yellow Fever, dengue, West Nile, and Zika viruses have high morbidity and mortality-about an estimated 20,000 deaths per year. As they depend on human vectors, they have expanded their geographical range in recent years due to altered climatic and social conditions. Despite these epidemiological and clinical premises, there are limited antiviral treatments for these infections. In this review, we describe the major compounds that are currently under evaluation for the treatment of flavivirus infections and the challenges faced during clinical trials, outlining their mechanisms of action in order to present an overview of ongoing studies. According to our review, the absence of approved antivirals for flaviviruses led to in vitro and in vivo experiments aimed at identifying compounds that can interfere with one or more viral cycle steps. Still, the currently unavailability of approved antivirals poses a significant public health issue.
Collapse
Affiliation(s)
- Erica Diani
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Anna Lagni
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Virginia Lotti
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
| | - Emil Tonon
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| | - Riccardo Cecchetto
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| | - Davide Gibellini
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, 37134 Verona, Italy; (A.L.); (V.L.); (R.C.)
- Unit of Microbiology, Azienda Ospedaliera Universitaria Integrata Verona, 37134 Verona, Italy;
| |
Collapse
|
30
|
Gupta N, Kodan P, Baruah K, Soneja M, Biswas A. Zika virus in India: past, present and future. QJM 2023; 116:644-649. [PMID: 31642501 DOI: 10.1093/qjmed/hcz273] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) is an arthropod-borne flavivirus that presents with acute febrile illness associated with rash, arthralgia and conjunctivitis. After years of sporadic reports in Africa, the three major outbreaks of this disease occurred in Yap Islands (2007), French Polynesia (2013-14) and South Americas (2015-16). Although, serological surveys suggested the presence of ZIKV in India in 1950s, cross-reactivity could not be ruled out. The first four proven cases of ZIKV from India were reported in 2017. This was followed by major outbreaks in the states of Rajasthan and Madhya Pradesh in 2018. Fortunately, the outbreaks in India were not associated with neurological complications. These outbreaks in India highlighted the spread of this disease beyond geographical barriers owing to the growing globalization, increased travel and ubiquitous presence of its vector, the Aedes mosquito. In this review, we discuss the epidemiology, clinical features and management of ZIKV in India.
Collapse
Affiliation(s)
- N Gupta
- From the Department of Medicine, All India Institute of Medical Sciences, 3rd Floor, Teaching Block, New Delhi 110029
| | - P Kodan
- From the Department of Medicine, All India Institute of Medical Sciences, 3rd Floor, Teaching Block, New Delhi 110029
| | - K Baruah
- National Vector Borne Disease Control Programme, Ministry of Health and Family Welfare, Government of India, 22 Shyam Nath Marg, New Delhi 110054, India
| | - M Soneja
- From the Department of Medicine, All India Institute of Medical Sciences, 3rd Floor, Teaching Block, New Delhi 110029
| | - A Biswas
- From the Department of Medicine, All India Institute of Medical Sciences, 3rd Floor, Teaching Block, New Delhi 110029
| |
Collapse
|
31
|
Wang L, Sanon A, Khoiriyah Z, Verwimp S, Abdelnabi R, Delang L. Tarsal exposure to atovaquone inhibits chikungunya virus transmission by Aedes aegypti mosquitoes, but not the transmission of Zika virus. Antiviral Res 2023; 217:105694. [PMID: 37532005 DOI: 10.1016/j.antiviral.2023.105694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/10/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
The antimalarial drug atovaquone was recently reported to inhibit the in vitro replication of different arboviruses, including chikungunya virus (CHIKV) and Zika virus (ZIKV). Furthermore, atovaquone was shown to block Plasmodium parasite transmission by Anopheles mosquitoes when the mosquitoes were exposed to low concentrations on treated surfaces (i.e. tarsal exposure). Therefore, we evaluated the anti-CHIKV and -ZIKV effects of atovaquone via tarsal exposure in Aedes aegypti mosquitoes. We first confirmed that atovaquone exerted a dose-dependent antiviral effect on CHIKV and ZIKV replication in mosquito-derived cells. The modest antiviral effect could be rescued by adding exogenous uridine. Next, we assessed the effect of tarsal exposure to atovaquone on the fitness of Ae. aegypti. Concentrations up to 100 μmol/m2 did not affect the fecundity and egg-hatching rate. No significant effect on mosquito survival was observed when mosquitoes were exposed to concentrations up to 25 μmol/m2. To evaluate the antiviral effect of atovaquone against CHIKV, we exposed female mosquitoes to 100 μmol/m2 atovaquone for 1h, after which the mosquitoes were immediately infected with CHIKV or ZIKV via bloodmeal. Atovaquone did not significantly reduce ZIKV or CHIKV infection in Ae. aegypti, but successfully blocked the transmission of CHIKV in saliva. Tarsal exposure to antiviral drugs could therefore be a potential new strategy to reduce virus transmission by mosquitoes.
Collapse
Affiliation(s)
- Lanjiao Wang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Aboubakar Sanon
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium; Laboratoire d'Entomologie Fondamentale et Appliquée, Université Joseph Ki-Zerbo, Burkina Faso
| | - Zakiyatul Khoiriyah
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium; Laboratory of Virology, Wageningen University and Research, 6708 PB, Wageningen, the Netherlands
| | - Sam Verwimp
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium
| | - Leen Delang
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
32
|
Bhat EA, Ali T, Sajjad N, Kumar R, Bron P. Insights into the structure, functional perspective, and pathogenesis of ZIKV: an updated review. Biomed Pharmacother 2023; 165:115175. [PMID: 37473686 DOI: 10.1016/j.biopha.2023.115175] [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: 06/02/2023] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 07/22/2023] Open
Abstract
Zika virus (ZIKV) poses a serious threat to the entire world. The rapid spread of ZIKV and recent outbreaks since 2007 have caused worldwide concern about the virus. Diagnosis is complicated because of the cross-reactivity of the virus with other viral antibodies. Currently, the virus is diagnosed by molecular techniques such as RT-PCR and IgM-linked enzyme immunoassays (MAC-ELISA). Recently, outbreaks and epidemics have been caused by ZIKV, and severe clinical symptoms and congenital malformations have also been associated with the virus. Although most ZIKV infections present with a subclinical or moderate flu-like course of illness, severe symptoms such as Guillain-Barre syndrome in adults and microcephaly in children of infected mothers have also been reported. Because there is no reliable cure for ZIKV and no vaccine is available, the public health response has focused primarily on preventing infection, particularly in pregnant women. A comprehensive approach is urgently needed to combat this infection and stop its spread and imminent threat. In view of this, this review aims to present the current structural and functional viewpoints, structure, etiology, clinical prognosis, and measures to prevent this transmission based on the literature and current knowledge. Moreover, we provide thorough description of the current understanding about ZIKV interaction with receptors, and a comparative examination of its similarities and differences with other viruses.
Collapse
Affiliation(s)
- Eijaz Ahmed Bhat
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, INSERM, 29 rue de Navacelles, 34090 Montpellier, France.
| | - Tufail Ali
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Nasreena Sajjad
- Department of Biochemistry, University of Kashmir, Hazratbal, Jammu and Kashmir 190006, India
| | - Rohit Kumar
- Department of Plant Molecular Biology, University of Delhi South Campus, New Delhi 110021, India
| | - Patrick Bron
- CBS (Centre de Biologie Structurale), Univ. Montpellier, CNRS, INSERM, 29 rue de Navacelles, 34090 Montpellier, France.
| |
Collapse
|
33
|
Chen Q, Li N, Zeng S, Wu S, Luo X, Zhang S, Zhu L, Wu J, Xie T, Bai S, Zhang H, Jiang Z, Lin S, Wu N, Jiang Y, Fang S, Wang X, Shu Y, Luo H. ZIKV infection differentially affects the transcriptional profiles in HTR8 and U251 cells. Virus Res 2023; 334:199166. [PMID: 37390859 PMCID: PMC10410584 DOI: 10.1016/j.virusres.2023.199166] [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: 12/29/2022] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
The mechanism by which Zika virus (ZIKV) causes severe birth defects in pregnant women remains unclear. Cell tropisms in placenta and brain play a crucial role in ZIKV pathogenesis, leading to congenital Zika syndrome (CZS). To identify the host factors involved in ZIKV infection, we compared the transcriptional profiles of ZIKV-infected human first-trimester placental trophoblast cells HTR8/SVneo and a human glioblastoma astrocytoma cell line U251. Our results demonstrated that ZIKV exhibited lower rates of mRNA replication and protein expression in HTR8 than in U251 cells, while showing a higher release of infectious viral particles. However, a greater number of differentially expressed genes (DEGs) were found in ZIKV-infected U251 cells than in ZIKV-infected HTR8 cells. Several of these DEGs were enriched in distinct biological processes related to the characteristics of each cell type that may contribute to foetal damage. Both cell types exhibited activation of common interferons, inflammatory cytokines, and chemokine production upon ZIKV infection. Moreover, the neutralization of tumour necrosis factor-alpha (TNF-α) promoted ZIKV infection in both trophoblasts and glioblastoma astrocytoma cells. Overall, we identified multiple DEGs associated with ZIKV pathogenesis.
Collapse
Affiliation(s)
- Qiqi Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Nina Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shike Zeng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Huizhou Municipal Central Hospital, Huizhou 516001, PR China
| | - Shu Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen 518172, PR China
| | - Xin Luo
- The Emergency Department, Eighth People's Hospital of Nanyang City, Nanyang 473000, PR China
| | - Shengze Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Lin Zhu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jiani Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Shaoxing Center for Disease Control and Prevention, Shaoxing 312075, PR China
| | - Ting Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shaohui Bai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Hao Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Zhiyuan Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shaoli Lin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Nan Wu
- Shenzhen Nanshan Center for Disease Control and Prevention, Shenzhen 518054, PR China
| | - Ying Jiang
- Shenzhen Nanshan Center for Disease Control and Prevention, Shenzhen 518054, PR China
| | - Shisong Fang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518073, PR China
| | - Xin Wang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518073, PR China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100176, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China.
| |
Collapse
|
34
|
de Siqueira Santos R, Rochael NC, Mattos TRF, Fallett E Silva MF, Linhares-Lacerda L, de Oliveira LT, Cunha MS, Mohana-Borges R, Gomes TA, Barbosa-Silva MC, Maron-Gutierrez T, Foguel D, Saraiva EM. Peripheral nervous system is injured by neutrophil extracellular traps (NETs) elicited by nonstructural (NS) protein-1 from Zika virus. FASEB J 2023; 37:e23126. [PMID: 37594040 DOI: 10.1096/fj.202201904r] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 07/05/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023]
Abstract
The involvement of innate immune mediators to the Zika virus (ZIKV)-induced neuroinflammation is not yet well known. Here, we investigated whether neutrophil extracellular traps (NETs), which are scaffolds of DNA associated with proteins, have the potential to injure peripheral nervous. The tissue lesions were evaluated after adding NETs to dorsal root ganglia (DRG) explants and to DRG constituent cells or injecting them into mouse sciatic nerves. Identification of NET harmful components was achieved by pharmacological inhibition of NET constituents. We found that ZIKV inoculation into sciatic nerves recruited neutrophils and elicited the production of the cytokines CXCL1 and IL-1β, classical NET inducers, but did not trigger NET formation. ZIKV blocked PMA- and CXCL8-induced NET release, but, in contrast, the ZIKV nonstructural protein (NS)-1 induced NET formation. NET-enriched supernatants were toxic to DRG explants, decreasing neurite area, length, and arborization. NETs were toxic to DRG constituent cells and affected myelinating cells. Myeloperoxidase (MPO) and histones were identified as the harmful component of NETs. NS1 injection into mouse sciatic nerves recruited neutrophils and triggered NET release and caspase-3 activation, events that were also elicited by the injection of purified MPO. In summary, we found that ZIKV NS1 protein induces NET formation, which causes nervous tissue damages. Our findings reveal new mechanisms leading to neuroinflammation by ZIKV.
Collapse
Affiliation(s)
- Raphael de Siqueira Santos
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Natalia Cadaxo Rochael
- Laboratório de Imunidade Inata, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Thayana Roberta F Mattos
- Laboratório de Imunidade Inata, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Matheus Felipe Fallett E Silva
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Leandra Linhares-Lacerda
- Laboratório de Imunidade Inata, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Leandro Teixeira de Oliveira
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Marcela Sabino Cunha
- Laboratório de Genética e Imunologia das Infecções Virais, Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ronaldo Mohana-Borges
- Laboratório de Biotecnologia e Bioengenharia Estrutural, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Tiago Araujo Gomes
- Laboratório de Microbiologia Celular Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Maria Carolina Barbosa-Silva
- Laboratório de Imunofarmacologia - Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Tatiana Maron-Gutierrez
- Laboratório de Imunofarmacologia - Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Debora Foguel
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Elvira Maria Saraiva
- Laboratório de Imunidade Inata, Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Shukla R, Chandra A, Kumar A, Kandpal P, Avashthi H, Goel VK, Qamar I, Singh N, Kelvin DJ, Singh TR. Repurposing of drugs against methyltransferase as potential Zika virus therapies. Sci Rep 2023; 13:7870. [PMID: 37188743 PMCID: PMC10184974 DOI: 10.1038/s41598-023-33341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
In recent years, the outbreak of infectious disease caused by Zika Virus (ZIKV) has posed a major threat to global public health, calling for the development of therapeutics to treat ZIKV disease. Several possible druggable targets involved in virus replication have been identified. In search of additional potential inhibitors, we screened 2895 FDA-approved compounds using Non-Structural Protein 5 (NS5) as a target utilizing virtual screening of in-silco methods. The top 28 compounds with the threshold of binding energy -7.2 kcal/mol value were selected and were cross-docked on the three-dimensional structure of NS5 using AutoDock Tools. Of the 2895 compounds screened, five compounds (Ceforanide, Squanavir, Amcinonide, Cefpiramide, and Olmesartan_Medoxomil) ranked highest based on filtering of having the least negative interactions with the NS5 and were selected for Molecular Dynamic Simulations (MDS) studies. Various parameters such as RMSD, RMSF, Rg, SASA, PCA and binding free energy were calculated to validate the binding of compounds to the target, ZIKV-NS5. The binding free energy was found to be -114.53, -182.01, -168.19, -91.16, -122.56, and -150.65 kJ mol-1 for NS5-SFG, NS5-Ceforanide, NS5-Squanavir, NS5-Amcinonide, NS5-Cefpiramide, and NS5-Ol_Me complexes respectively. The binding energy calculations suggested Cefpiramide and Olmesartan_Medoxomil (Ol_Me) as the most stable compounds for binding to NS5, indicating a strong rationale for their use as lead compounds for development of ZIKV inhibitors. As these drugs have been evaluated on pharmacokinetics and pharmacodynamics parameters only, in vitro and in vivo testing and their impact on Zika viral cell culture may suggest their clinical trials on ZIKV patients.
Collapse
Affiliation(s)
- Rohit Shukla
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India
| | - Anshuman Chandra
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India
- School of Physical Science, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Anuj Kumar
- Laboratory of Immunity, Shantou University Medical College, Shantou, China
- Department of Microbiology and Immunology, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada
- Department of Pediatrics, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada
- European Virus Bioinformatics Center, Leutragraben 1, Jena, Germany
| | | | - Himanshu Avashthi
- Division of Agricultural Bioinformatics, ICAR-Indian Agricultural Statistics Research Institute, Pusa, New Delhi, India
| | - Vijay Kumar Goel
- School of Physical Science, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Imteyaz Qamar
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India
| | - Nagendra Singh
- School of Biotechnology, Gautam Buddha University, Gautam Buddh Nagar, Greater Noida, Uttar Pradesh, 201312, India.
| | - David J Kelvin
- Laboratory of Immunity, Shantou University Medical College, Shantou, China.
- Department of Microbiology and Immunology, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada.
- Department of Pediatrics, IWK Health Center, Canadian Centre for Vaccinology CCfV, Faculty of Medicine, Dalhousie University, Halifax, Canada.
| | - Tiratha Raj Singh
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India.
- Centre for Excellence in Healthcare Technologies and Informatics (CEHTI), Jaypee University of Information Technology (JUIT), Waknaghat, Solan, Himachal Pradesh, 173234, India.
| |
Collapse
|
36
|
Frank JC, Song BH, Lee YM. Mice as an Animal Model for Japanese Encephalitis Virus Research: Mouse Susceptibility, Infection Route, and Viral Pathogenesis. Pathogens 2023; 12:pathogens12050715. [PMID: 37242385 DOI: 10.3390/pathogens12050715] [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: 03/11/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Japanese encephalitis virus (JEV), a zoonotic flavivirus, is principally transmitted by hematophagous mosquitoes, continually between susceptible animals and incidentally from those animals to humans. For almost a century since its discovery, JEV was geographically confined to the Asia-Pacific region with recurrent sizable outbreaks involving wildlife, livestock, and people. However, over the past decade, it has been detected for the first time in Europe (Italy) and Africa (Angola) but has yet to cause any recognizable outbreaks in humans. JEV infection leads to a broad spectrum of clinical outcomes, ranging from asymptomatic conditions to self-limiting febrile illnesses to life-threatening neurological complications, particularly Japanese encephalitis (JE). No clinically proven antiviral drugs are available to treat the development and progression of JE. There are, however, several live and killed vaccines that have been commercialized to prevent the infection and transmission of JEV, yet this virus remains the main cause of acute encephalitis syndrome with high morbidity and mortality among children in the endemic regions. Therefore, significant research efforts have been directed toward understanding the neuropathogenesis of JE to facilitate the development of effective treatments for the disease. Thus far, multiple laboratory animal models have been established for the study of JEV infection. In this review, we focus on mice, the most extensively used animal model for JEV research, and summarize the major findings on mouse susceptibility, infection route, and viral pathogenesis reported in the past and present, and discuss some unanswered key questions for future studies.
Collapse
Affiliation(s)
- Jordan C Frank
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Byung-Hak Song
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| | - Young-Min Lee
- Department of Animal, Dairy, and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA
| |
Collapse
|
37
|
Rosado LEP, Martelli CMT, Brickley EB, Gomes MBF, de Toledo Lima T, da Costa PSS, de Ávila MP, Viggiano MB, do Amaral WN, de Rezende Feres VC, Fiaccadori FS, de Sene Amancio Zara AL, Ferreira-Lopes A, Turchi MD. Risk of adverse pregnancy and infant outcomes associated with prenatal Zika virus infection: a post-epidemic cohort in Central-West Brazil. Sci Rep 2023; 13:7335. [PMID: 37147405 PMCID: PMC10161159 DOI: 10.1038/s41598-023-33334-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/11/2023] [Indexed: 05/07/2023] Open
Abstract
This study aimed to estimate the risks of adverse infant outcomes in the first year of life related to prenatal Zika virus (ZIKV) exposure. A prospective cohort of pregnant women with rash was recruited in Central-West Brazil in a post-epidemic period (January 2017 to April 2019). We evaluated participants' medical histories and performed ZIKV diagnostic testing using molecular (reverse transcription polymerase chain reaction [RT-PCR]) and serologic (immunoglobulin [Ig]M and plaque reduction neutralization tests [PRNT90]) assays. The ZIKV-positive group included both RT-PCR-confirmed cases as well as IgM and/or PRNT90-positive probable cases. Children were evaluated at birth and in the first 12 months of life. Transfontanellar ultrasound, central nervous system computed tomography, eye fundoscopy and retinography were performed. We estimated the absolute risk and 95% confidence interval (95% CI) of adverse infant outcomes among confirmed prenatally ZIKV-exposed children. Among 81 pregnant women with rash, 43 (53.1%) were ZIKV infected. The absolute risk of microcephaly among offspring of ZIKV-infected pregnant women was 7.0% (95% CI: 1.5-19.1), including the two cases of microcephaly detected prenatally and one detected postnatally. In total, 54.5% (95% CI: 39.8-68.7) of children in the ZIKV-exposed group had at least one ophthalmic abnormality, with the most frequent abnormalities being focal pigmentary mottling and chorioretinal atrophy or scarring. Our findings reinforce the importance of long-term monitoring of prenatally ZIKV-exposed children born apparently asymptomatic for Congenital Zika Syndrome.
Collapse
Affiliation(s)
- Luiza Emylce Pela Rosado
- Graduate Program in Tropical Medicine and Public Health of the Federal University of Goias, Goiânia, Brazil
- Obstetrics Department, Maternal and Infant Hospital of Goias State, Goiânia, Brazil
| | | | - Elizabeth B Brickley
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Maria Barbara Franco Gomes
- Graduate Program in Health Sciences of the Federal University of Goias, Goiânia, Brazil
- Pediatric Department, Maternal and Infant Hospital of Goias State, Goiânia, Brazil
| | - Talita de Toledo Lima
- Graduate Program in Health Sciences of the Federal University of Goias, Goiânia, Brazil
- Reference Center in Ophthalmology of the Federal University of Goias, Goiânia, Brazil
| | | | - Marcos Pereira de Ávila
- Reference Center in Ophthalmology of the Federal University of Goias, Goiânia, Brazil
- Retina and Vitreous Department, School of Medicine, Federal University of Goias, Goiânia, Brazil
| | | | | | | | - Fabiola Souza Fiaccadori
- Virology Department, Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Brazil
| | | | | | - Marilia Dalva Turchi
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Brazil.
| |
Collapse
|
38
|
Nazneen F, Thompson EA, Blackwell C, Bai JS, Huang F, Bai F. An effective live-attenuated Zika vaccine candidate with a modified 5' untranslated region. NPJ Vaccines 2023; 8:50. [PMID: 37005424 PMCID: PMC10066991 DOI: 10.1038/s41541-023-00650-w] [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: 08/15/2022] [Accepted: 03/17/2023] [Indexed: 04/04/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted flavivirus that has caused devastating congenital Zika syndrome (CZS), including microcephaly, congenital malformation, and fetal demise in human newborns in recent epidemics. ZIKV infection can also cause Guillain-Barré syndrome (GBS) and meningoencephalitis in adults. Despite intensive research in recent years, there are no approved vaccines or antiviral therapeutics against CZS and adult Zika diseases. In this report, we developed a novel live-attenuated ZIKV strain (named Z7) by inserting 50 RNA nucleotides (nt) into the 5' untranslated region (UTR) of a pre-epidemic ZIKV Cambodian strain, FSS13025. We used this particular ZIKV strain as it is attenuated in neurovirulence, immune antagonism, and mosquito infectivity compared with the American epidemic isolates. Our data demonstrate that Z7 replicates efficiently and produces high titers without causing apparent cytopathic effects (CPE) in Vero cells or losing the insert sequence, even after ten passages. Significantly, Z7 induces robust humoral and cellular immune responses that completely prevent viremia after a challenge with a high dose of an American epidemic ZIKV strain PRVABC59 infection in type I interferon (IFN) receptor A deficient (Ifnar1-/-) mice. Moreover, adoptive transfer of plasma collected from Z7 immunized mice protects Ifnar1-/- mice from ZIKV (strain PRVABC59) infection. These results suggest that modifying the ZIKV 5' UTR is a novel strategy to develop live-attenuated vaccine candidates for ZIKV and potentially for other flaviviruses.
Collapse
Affiliation(s)
- Farzana Nazneen
- Cell and Molecular Biology Program, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - E Ashley Thompson
- Cell and Molecular Biology Program, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Claire Blackwell
- Cell and Molecular Biology Program, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Jonathan S Bai
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Faqing Huang
- Chemistry and Biochemistry Program, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, 39406, USA
| | - Fengwei Bai
- Cell and Molecular Biology Program, Center for Molecular and Cellular Biosciences, The University of Southern Mississippi, Hattiesburg, MS, 39406, USA.
| |
Collapse
|
39
|
Martinez-Cruz C, Arenas-Monreal L, Gomez-Dantes H, Villegas-Chim J, Barrera-Fuentes Gloria A, Toledo-Romani Maria E, Pavia-Ruz N, Che-Mendoza A, Manrique-Saide P. Educational intervention for the control of Aedes aegypti with Wolbachia in Yucatan, Mexico. EVALUATION AND PROGRAM PLANNING 2023; 97:102205. [PMID: 36580820 DOI: 10.1016/j.evalprogplan.2022.102205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/16/2022] [Accepted: 12/13/2022] [Indexed: 06/17/2023]
Abstract
UNLABELLED The implementation of new control strategies for Aedes aegypti (Ae. Aegpyti), a vector of dengue, chikungunya, and Zika viruses, requires communities to adopt specific behaviors to achieve the success of these innovations. AIM We evaluated the effect of an educational intervention based on the Precede-Proceed Model (PPM) and the Diffusion of Innovations Theory (DIT) for the control and prevention of diseases transmitted by Ae. aegypti through release of male mosquitoes infected with Wolbachia bacteria in a suburban town in Yucatan, Mexico. MATERIAL AND METHODS From July 2019 to February 2020, a quasi-experimental study was carried out through an educational intervention (pre- and post-measurements) using quantitative-qualitative techniques, in a Yucatan suburban town where male mosquitoes with Wolbachia were released for the suppression of Ae. aegypti populations. Eleven educational workshops were attended by heads of household (n = 19) and schoolchildren (n = 11). Other 136 heads of household not attending the workshops received information individually. RESULTS The educational intervention had a significant effect on the mean scores of the contributing and behavioral factors for adoption of innovation (p < 0.05) in the pre- and post-intervention measurements. CONCLUSION Innovative methods for the control and prevention of diseases related to Aedes aegypti can be strengthened through educational interventions supported by sound methodologies. DESCRIPTORS Community health education, Aedes aegypti, Wolbachia, Mexico.
Collapse
Affiliation(s)
- Carolina Martinez-Cruz
- School of Public Health of Mexico/National Institute of Public Health, University No. 655 Colonia Santa María Ahuacatitlán, Los Pinos and Caminera, Cuernavaca, Morelos, Mexico.
| | - Luz Arenas-Monreal
- Health Systems Research Center/National Institute of Health, University No. 655 Colonia Santa María Ahuacatitlán, Los Pinos and Caminera, Cuernavaca, Morelos, Mexico.
| | - Héctor Gomez-Dantes
- Health Systems Research Center/National Institute of Health, University No. 655 Colonia Santa María Ahuacatitlán, Los Pinos and Caminera, Cuernavaca, Morelos, Mexico.
| | - Josue Villegas-Chim
- Collaborative Unit for Entomological Bioassays, Campus of Biological and Agricultural Sciences, Autonomous University of Yucatan, Carretera Merida-Xmatkuil Km. 15.5 Apdo., Plan de Ayala II, Itzimná, 97100 Mérida, Yucatan, Mexico,.
| | - Abigail Barrera-Fuentes Gloria
- Collaborative Unit for Entomological Bioassays, Campus of Biological and Agricultural Sciences, Autonomous University of Yucatan, Carretera Merida-Xmatkuil Km. 15.5 Apdo., Plan de Ayala II, Itzimná, 97100 Mérida, Yucatan, Mexico,.
| | - Eugenia Toledo-Romani Maria
- Pedro Kouri Institute of Tropical Medicine, Avenida Novia del Mediodia, KM 6 1/2, La Lisa, Havana 11400, Cuba.
| | - Norma Pavia-Ruz
- Regional Research Center, Biomedical Unit, Autonomous University of Yucatan, Av. Itzáes, Centro, 97000 Mérida, Yucatan, Mexico.
| | - Azael Che-Mendoza
- Collaborative Unit for Entomological Bioassays, Campus of Biological and Agricultural Sciences, Autonomous University of Yucatan, Carretera Merida-Xmatkuil Km. 15.5 Apdo., Plan de Ayala II, Itzimná, 97100 Mérida, Yucatan, Mexico,.
| | - Pablo Manrique-Saide
- Collaborative Unit for Entomological Bioassays, Campus of Biological and Agricultural Sciences, Autonomous University of Yucatan, Carretera Merida-Xmatkuil Km. 15.5 Apdo., Plan de Ayala II, Itzimná, 97100 Mérida, Yucatan, Mexico,.
| |
Collapse
|
40
|
Huang S, Wang J, Xiong Y, Liu C, Qi Y, Zou K, Tan J, Sun X. Impact of maternal hepatitis B carrier status on congenital abnormalities: a systematic review and meta-analysis. BMJ Open 2023; 13:e066017. [PMID: 36977541 PMCID: PMC10069551 DOI: 10.1136/bmjopen-2022-066017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVES This study aims to explore whether maternal hepatitis B carrier status is associated with an increased risk of congenital abnormalities. DESIGN A systematic review and meta-analysis of observational studies. DATA SOURCES PubMed, Embase (Ovid), Scopus, the China National Knowledge Infrastructure (CNKI) and the Wanfang databases. STUDY SELECTION Five databases were searched systematically from inception to 7 September 2021. Cohort and case-control studies that investigated the association between maternal hepatitis B virus (HBV) infection and congenital abnormalities were included. This study was conducted according to MOOSE (Meta-analysis of Observational Studies in Epidemiology) guidelines. DATA EXTRACTION AND SYNTHESIS Two reviewers independently collected data, as well as assessed risk of bias by using Newcastle-Ottawa Scale. We pooled crude relative risk (cRR) and adjusted OR (aOR) by DerSimonian-Laird random-effects model. Heterogeneity was explored by I 2 statistics, Cochran's Q test. Several subgroup analyses and sensitivity analyses were performed. RESULTS In total, 14 studies involving 16 205 pregnant women exposed to HBV were included. The pooled cRR of 1.15 (95% CI: 0.92 to 1.45; 14 studies included) showed a marginal but not significant association between maternal HBV-carrier status and congenital abnormalities. However, the pooled aOR of 1.40 (95% CI: 1.01 to 1.93; 8 studies included) indicated that pregnant women with HBV infection might be associated with a higher risk of congenital abnormalities. Subgroup analyses of adjusted data showed a higher pooling cRR or aOR on high prevalence HBV infection populations, as well as studies from Asia and Oceania. CONCLUSIONS Maternal hepatitis B carrier status might be at potential risk for congenital abnormalities. The existing evidence was not sufficient to draw a firm conclusion. Additional studies may be warranted to confirm the association. PROSPERO REGISTRATION NUMBER CRD42020205459.
Collapse
Affiliation(s)
- Shiyao Huang
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
- Sichuan Evidence-based Medicine Center of Traditional Chinese Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jing Wang
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiquan Xiong
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| | - Chunrong Liu
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| | - Yana Qi
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| | - Kang Zou
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| | - Jing Tan
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| | - Xin Sun
- Chinese Evidence-based Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- NMPA Key Laboratory for Real World Data Research and Evaluation in Hainan, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Dias SSG, Cunha-Fernandes T, Souza-Moreira L, Soares VC, Lima GB, Azevedo-Quintanilha IG, Santos J, Pereira-Dutra F, Freitas C, Reis PA, Rehen SK, Bozza FA, Souza TML, de Almeida CJG, Bozza PT. Metabolic reprogramming and lipid droplets are involved in Zika virus replication in neural cells. J Neuroinflammation 2023; 20:61. [PMID: 36882750 PMCID: PMC9992922 DOI: 10.1186/s12974-023-02736-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Zika virus (ZIKV) infection is a global public health concern linked to adult neurological disorders and congenital diseases in newborns. Host lipid metabolism, including lipid droplet (LD) biogenesis, has been associated with viral replication and pathogenesis of different viruses. However, the mechanisms of LD formation and their roles in ZIKV infection in neural cells are still unclear. Here, we demonstrate that ZIKV regulates the expression of pathways associated with lipid metabolism, including the upregulation and activation of lipogenesis-associated transcription factors and decreased expression of lipolysis-associated proteins, leading to significant LD accumulation in human neuroblastoma SH-SY5Y cells and in neural stem cells (NSCs). Pharmacological inhibition of DGAT-1 decreased LD accumulation and ZIKV replication in vitro in human cells and in an in vivo mouse model of infection. In accordance with the role of LDs in the regulation of inflammation and innate immunity, we show that blocking LD formation has major roles in inflammatory cytokine production in the brain. Moreover, we observed that inhibition of DGAT-1 inhibited the weight loss and mortality induced by ZIKV infection in vivo. Our results reveal that LD biogenesis triggered by ZIKV infection is a crucial step for ZIKV replication and pathogenesis in neural cells. Therefore, targeting lipid metabolism and LD biogenesis may represent potential strategies for anti-ZIKV treatment development.
Collapse
Affiliation(s)
- Suelen Silva Gomes Dias
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Tamires Cunha-Fernandes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Luciana Souza-Moreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Programa de Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle Barbosa Lima
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Julia Santos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Filipe Pereira-Dutra
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Caroline Freitas
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Patricia A Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil.,Instituto de Biologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil.,Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro, Brazil
| | - Thiago M Lopes Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Inovação em Doenças de Populações Negligenciadas (INCT/IDPN), Centro de Desenvolvimento Tecnológico em Saúde, (CDTS), FIOCRUZ, Rio de Janeiro, Brazil
| | - Cecilia J G de Almeida
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Patricia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.
| |
Collapse
|
42
|
Astrocytes in the pathophysiology of neuroinfection. Essays Biochem 2023; 67:131-145. [PMID: 36562155 DOI: 10.1042/ebc20220082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/24/2022]
Abstract
Key homeostasis providing cells in the central nervous system (CNS) are astrocytes, which belong to the class of cells known as atroglia, a highly heterogeneous type of neuroglia and a prominent element of the brain defence. Diseases evolve due to altered homeostatic state, associated with pathology-induced astroglia remodelling represented by reactive astrocytes, astroglial atrophy and astrodegeneration. These features are hallmarks of most infectious insults, mediated by bacteria, protozoa and viruses; they are also prominent in the systemic infection. The COVID-19 pandemic revived the focus into neurotropic viruses such as SARS-CoV2 (Coronaviridae) but also the Flaviviridae viruses including tick-borne encephalitis (TBEV) and Zika virus (ZIKV) causing the epidemic in South America prior to COVID-19. Astrocytes provide a key response to neurotropic infections in the CNS. Astrocytes form a parenchymal part of the blood-brain barrier, the site of virus entry into the CNS. Astrocytes exhibit aerobic glycolysis, a form of metabolism characteristic of highly morphologically plastic cells, like cancer cells, hence a suitable milieu for multiplication of infectious agent, including viral particles. However, why the protection afforded by astrocytes fails in some circumstances is an open question to be studied in the future.
Collapse
|
43
|
Santos ES, Miranda JG, Saba H, Skalinski LM, Araújo ML, Veiga RV, Costa MDCN, Cardim LL, Paixão ES, Teixeira MG, Andrade RF, Barreto ML. Complex network analysis of arboviruses in the same geographic domain: Differences and similarities. CHAOS, SOLITONS, AND FRACTALS 2023; 168:None. [PMID: 36876054 PMCID: PMC9980430 DOI: 10.1016/j.chaos.2023.113134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/29/2022] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
Arbovirus can cause diseases with a broad spectrum from mild to severe and long-lasting symptoms, affecting humans worldwide and therefore considered a public health problem with global and diverse socio-economic impacts. Understanding how they spread within and across different regions is necessary to devise strategies to control and prevent new outbreaks. Complex network approaches have widespread use to get important insights on several phenomena, as the spread of these viruses within a given region. This work uses the motif-synchronization methodology to build time varying complex networks based on data of registered infections caused by Zika, chikungunya, and dengue virus from 2014 to 2020, in 417 cities of the state of Bahia, Brazil. The resulting network sets capture new information on the spread of the diseases that are related to the time delay in the synchronization of the time series among different municipalities. Thus the work adds new and important network-based insights to previous results based on dengue dataset in the period 2001-2016. The most frequent synchronization delay time between time series in different cities, which control the insertion of edges in the networks, ranges 7 to 14 days, a period that is compatible with the time of the individual-mosquito-individual transmission cycle of these diseases. As the used data covers the initial periods of the first Zika and chikungunya outbreaks, our analyses reveal an increasing monotonic dependence between distance among cities and the time delay for synchronization between the corresponding time series. The same behavior was not observed for dengue, first reported in the region back in 1986, either in the previously 2001-2016 based results or in the current work. These results show that, as the number of outbreaks accumulates, different strategies must be adopted to combat the dissemination of arbovirus infections.
Collapse
Affiliation(s)
- Eslaine S. Santos
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - José G.V. Miranda
- Physics Institute, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Hugo Saba
- Centro Universitário SENAI CIMATEC, Av. Orlando Gomes, 1845—Piatã, Salvador 41650-010, Brazil
- Department of Exact and Earth Sciences, University of the State of Bahia, R. Silveira Martins, 2555—Cabula, Salvador 41180-045, Brazil
| | - Lacita M. Skalinski
- Collective Health Institute, Federal University of Bahia, Salvador, Bahia, Brazil
- Santa Cruz State University, Ilhéus, Bahia, Brazil
| | - Marcio L.V. Araújo
- Instituto Federal de Ciência e Tecnologia da Bahia (IFBA), R. São Cristóvão, s/n - Novo Horizonte, Lauro de Freitas, 42700-000, Brazil
| | - Rafael V. Veiga
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
- The Babraham Institute, Laboratory of Lymphocyte Signalling and Development, Cambridge, United Kingdom
| | | | - Luciana L. Cardim
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
| | - Enny S. Paixão
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Maria Glória Teixeira
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
- Collective Health Institute, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Roberto F.S. Andrade
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
- Physics Institute, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Maurício L. Barreto
- Center of Data and Knowledge Integration for Health (CIDACS), Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, Brazil
- Collective Health Institute, Federal University of Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
44
|
In Vitro and In Vivo Coinfection and Superinfection Dynamics of Mayaro and Zika Viruses in Mosquito and Vertebrate Backgrounds. J Virol 2023; 97:e0177822. [PMID: 36598200 PMCID: PMC9888278 DOI: 10.1128/jvi.01778-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Globalization and climate change have contributed to the simultaneous increase and spread of arboviral diseases. Cocirculation of several arboviruses in the same geographic region provides an impetus to study the impacts of multiple concurrent infections within an individual vector mosquito. Here, we describe coinfection and superinfection with the Mayaro virus (Togaviridae, Alphavirus) and Zika virus (Flaviviridae, Flavivirus) in vertebrate and mosquito cells, as well as Aedes aegypti adult mosquitoes, to understand the interaction dynamics of these pathogens and effects on viral infection, dissemination, and transmission. Aedes aegypti mosquitoes were able to be infected with and transmit both pathogens simultaneously. However, whereas Mayaro virus was largely unaffected by coinfection, it had a negative impact on infection and dissemination rates for Zika virus compared to single infection scenarios. Superinfection of Mayaro virus atop a previous Zika virus infection resulted in increased Mayaro virus infection rates. At the cellular level, we found that mosquito and vertebrate cells were also capable of being simultaneously infected with both pathogens. Similar to our findings in vivo, Mayaro virus negatively affected Zika virus replication in vertebrate cells, displaying complete blocking under certain conditions. Viral interference did not occur in mosquito cells. IMPORTANCE Epidemiological and clinical studies indicate that multiple arboviruses are cocirculating in human populations, leading to some individuals carrying more than one arbovirus at the same time. In turn, mosquitoes can become infected with multiple pathogens simultaneously (coinfection) or sequentially (superinfection). Coinfection and superinfection can have synergistic, neutral, or antagonistic effects on viral infection dynamics and ultimately have impacts on human health. Here we investigate the interaction between Zika virus and Mayaro virus, two emerging mosquito-borne pathogens currently circulating together in Latin America and the Caribbean. We find a major mosquito vector of these viruses-Aedes aegypti-can carry and transmit both arboviruses at the same time. Our findings emphasize the importance of considering co- and superinfection dynamics during vector-pathogen interaction studies, surveillance programs, and risk assessment efforts in epidemic areas.
Collapse
|
45
|
Safadi DE, Lebeau G, Lagrave A, Mélade J, Grondin L, Rosanaly S, Begue F, Hoareau M, Veeren B, Roche M, Hoarau JJ, Meilhac O, Mavingui P, Desprès P, Viranaïcken W, Krejbich-Trotot P. Extracellular Vesicles Are Conveyors of the NS1 Toxin during Dengue Virus and Zika Virus Infection. Viruses 2023; 15:v15020364. [PMID: 36851578 PMCID: PMC9965858 DOI: 10.3390/v15020364] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Extracellular vesicles (EVs), produced during viral infections, are of emerging interest in understanding infectious processes and host-pathogen interactions. EVs and exosomes in particular have the natural ability to transport nucleic acids, proteins, and other components of cellular or viral origin. Thus, they participate in intercellular communication, immune responses, and infectious and pathophysiological processes. Some viruses are known to hijack the cell production and content of EVs for their benefit. Here, we investigate whether two pathogenic flaviviruses i.e., Zika Virus (ZIKV) and Dengue virus (DENV2) could have an impact on the features of EVs. The analysis of EVs produced by infected cells allowed us to identify that the non-structural protein 1 (NS1), described as a viral toxin, is associated with exosomes. This observation could be confirmed under conditions of overexpression of recombinant NS1 from each flavivirus. Using different isolation methods (i.e., exosome isolation kit, size exclusion chromatography, Polyethylene Glycol enrichment, and ELISA capture), we showed that NS1 was present as a dimer at the surface of excreted exosomes, and that this association could occur in the extracellular compartment. This finding could be of major importance in a physiological context. Indeed, this capacity of NS1 to address EVs and its implication in the pathophysiology during Dengue or Zika diseases should be explored. Furthermore, exosomes that have demonstrated a natural capacity to vectorize NS1 could serve as useful tools for vaccine development.
Collapse
Affiliation(s)
- Daed El Safadi
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Grégorie Lebeau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Alisé Lagrave
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Julien Mélade
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Lauriane Grondin
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Sarah Rosanaly
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Floran Begue
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Mathilde Hoareau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Bryan Veeren
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Marjolaine Roche
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Jean-Jacques Hoarau
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Olivier Meilhac
- Unité Mixte Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), Université de la Réunion, INSERM, UMR 1188, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Patrick Mavingui
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Philippe Desprès
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
| | - Wildriss Viranaïcken
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
- Correspondence: (W.V.); (P.K.-T.)
| | - Pascale Krejbich-Trotot
- Unité Mixte Processus Infectieux en Milieu Insulaire Tropical (PIMIT), Université de la Réunion, INSERM U1187, CNRS UMR 9192, IRD UMR 249, Plateforme Technologique CYROI, 97490 Saint-Denis de La Réunion, France
- Correspondence: (W.V.); (P.K.-T.)
| |
Collapse
|
46
|
Kozyreva AA, Bembeeva RT, Druzhinina ES, Zavadenko NN. [Guillain-Barre syndrome in children]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:20-32. [PMID: 37942969 DOI: 10.17116/jnevro202312309220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Guillain-Barré syndrome (GBS) is an immune-mediated disease of the peripheral nervous system that can occur in both children and adults. The classic presentation of GBS is characterized by progressive symmetrical, ascending muscle weakness. Patients with GBS require meticulous monitoring due to the risk of bulbar syndrome, respiratory failure and autonomic dysfunction, which can be life-threatening. Early diagnosis and timely prescription of pathogenetic therapy for GBS are particularly important, especially in young children. Meanwhile, the spectrum of disorders covered by GBS has expanded significantly; its eponym is now designate any variant of acute dysimmune polyneuropathy, and its atypical forms pose a serious diagnostic problem for clinicians. This review article provides an analysis of the data available in the medical literature on GBS in children and discusses the tactics for diagnosing and managing patients with GBS, taking into account the Russian and European clinical recommendations.
Collapse
Affiliation(s)
- A A Kozyreva
- Pirogov Russian National Research Medical University, Moscow, Russia
- Russian Children's Clinical Hospital, Moscow, Russia
| | - R Ts Bembeeva
- Pirogov Russian National Research Medical University, Moscow, Russia
- Russian Children's Clinical Hospital, Moscow, Russia
| | - E S Druzhinina
- Pirogov Russian National Research Medical University, Moscow, Russia
- Russian Children's Clinical Hospital, Moscow, Russia
| | - N N Zavadenko
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
47
|
Kadawathagedara M, Muckle G, Quénel P, Michineau L, Le Bot B, Hoen B, Tressieres B, Multigner L, Chevrier C, Cordier S. Infant neurodevelopment and behavior in Guadeloupe after lead exposure and Zika maternal infection during pregnancy. Neurotoxicology 2023; 94:135-146. [PMID: 36402195 DOI: 10.1016/j.neuro.2022.11.007] [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: 08/03/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Prenatal lead exposure is known to have neurotoxic effects on the developing fetus, while some viral infections may have a tropism for the central nervous system. Our objective was to study whether the effects of prenatal lead exposure on infant development and behaviors at 18 months of age are modified by the occurrence of a maternal infection to Zika virus (ZIKV) during pregnancy. METHODS During the ZIKV epidemic in Guadeloupe in 2016 a cohort of pregnant women was set up. Blood samples (pregnancy, childbirth and cord) (n = 297) enabled us to measure blood lead levels aimed to determine prenatal lead exposure and the likelihood of maternal infection during pregnancy (ZIKV status + vs -). The 18 months "Ages and Stages Questionnaire" (ASQ) was used to generate scores for global development, fine and gross motor skills, communication, problem solving, and personal-social skills. The questions from a longitudinal cohort study conducted in Canada (Québec) were used to generate hyperactivity, opposition, inattention and physical aggression scores. Associations were tested by multivariate linear regressions. RESULTS Prenatal lead exposure was associated with delays in neurodevelopment at 18 months, reflected by lower scores in ASQ totals, and in the fine motor and problem-solving domains. Some of these associations appeared to be sex-specific, observed almost exclusively in boys (ASQ total, fine motor and personal-social scores). Prenatal lead exposure was not associated with behavioral scores. ZIKV infection during pregnancy was associated with a lower fine motor ASQ score, and higher scores for hyperactivity, opposition and physical aggression. Significant interaction between prenatal lead exposure and ZIKV status was observed with a lower personal-social score in ZIKV (-) only, and for hyperactivity and inattention scores, though some of these interactions (ASQ personal-social score, inattention score) were no longer significant when children with microcephaly were excluded from the analyses. DISCUSSION/CONCLUSION Our study confirms previous findings of associations between prenatal exposure to lead at low levels and adverse neurodevelopmental outcomes during infancy and the particular vulnerability of boys. It suggests associations between ZIKV infection during pregnancy and adverse effects on a number of neurodevelopmental functions (fine motor function) and behaviors (opposition, hyperactivity), that need to be confirmed at later age. There is no strong evidence of interaction between ZIKV infection and lead exposure but both prenatal risk factors may affect fine motor function.
Collapse
Affiliation(s)
- M Kadawathagedara
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - G Muckle
- École de psychologie, Université Laval, et Centre de recherche du CHU de Québec-Université Laval, Québec, Canada
| | - P Quénel
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - L Michineau
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - B Le Bot
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - B Hoen
- Centre d'Investigation Clinique Antilles Guyane CIC 1424 Inserm, CHU de Guadeloupe, 97159 Pointe-à-Pitre, France
| | - B Tressieres
- Centre d'Investigation Clinique Antilles Guyane CIC 1424 Inserm, CHU de Guadeloupe, 97159 Pointe-à-Pitre, France
| | - L Multigner
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - C Chevrier
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France
| | - S Cordier
- Univ Rennes, EHESP, Inserm, IRSET UMR_S 1085, F-35000 Rennes, France.
| |
Collapse
|
48
|
Eder J, Zijlstra-Willems E, Koen G, Kootstra NA, Wolthers KC, Geijtenbeek TB. Transmission of Zika virus by dendritic cell subsets in skin and vaginal mucosa. Front Immunol 2023; 14:1125565. [PMID: 36949942 PMCID: PMC10025456 DOI: 10.3389/fimmu.2023.1125565] [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: 12/16/2022] [Accepted: 02/23/2023] [Indexed: 03/08/2023] Open
Abstract
Zika virus is a member of the Flaviviridae family that has caused recent outbreaks associated with neurological malformations. Transmission of Zika virus occurs primarily via mosquito bite but also via sexual contact. Dendritic cells (DCs) and Langerhans cells (LCs) are important antigen presenting cells in skin and vaginal mucosa and paramount to induce antiviral immunity. To date, little is known about the first cells targeted by Zika virus in these tissues as well as subsequent dissemination of the virus to other target cells. We therefore investigated the role of DCs and LCs in Zika virus infection. Human monocyte derived DCs (moDCs) were isolated from blood and primary immature LCs were obtained from human skin and vaginal explants. Zika virus exposure to moDCs but not skin and vaginal LCs induced Type I Interferon responses. Zika virus efficiently infected moDCs but neither epidermal nor vaginal LCs became infected. Infection of a human full skin model showed that DC-SIGN expressing dermal DCs are preferentially infected over langerin+ LCs. Notably, not only moDCs but also skin and vaginal LCs efficiently transmitted Zika virus to target cells. Transmission by LCs was independent of direct infection of LCs. These data suggest that DCs and LCs are among the first target cells for Zika virus not only in the skin but also the genital tract. The role of vaginal LCs in dissemination of Zika virus from the vaginal mucosa further emphasizes the threat of sexual transmission and supports the investigation of prophylaxes that go beyond mosquito control.
Collapse
Affiliation(s)
- Julia Eder
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Esther Zijlstra-Willems
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Gerrit Koen
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Neeltje A. Kootstra
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Katja C. Wolthers
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
| | - Teunis B. Geijtenbeek
- Department of Experimental Immunology, Amsterdam UMC location University of Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
- *Correspondence: Teunis B. Geijtenbeek,
| |
Collapse
|
49
|
Modeling the spread of the Zika virus by sexual and mosquito transmission. PLoS One 2022; 17:e0270127. [PMID: 36584063 PMCID: PMC9803243 DOI: 10.1371/journal.pone.0270127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 06/05/2022] [Indexed: 12/31/2022] Open
Abstract
Zika Virus (ZIKV) is a flavivirus that is transmitted predominantly by the Aedes species of mosquito, but also through sexual contact, blood transfusions, and congenitally from mother to child. Although approximately 80% of ZIKV infections are asymptomatic and typical symptoms are mild, multiple studies have demonstrated a causal link between ZIKV and severe diseases such as Microcephaly and Guillain Barré Syndrome. Two goals of this study are to improve ZIKV models by considering the spread dynamics of ZIKV as both a vector-borne and sexually transmitted disease, and also to approximate the degree of under-reporting. In order to accomplish these objectives, we propose a compartmental model that allows for the analysis of spread dynamics as both a vector-borne and sexually transmitted disease, and fit it to the ZIKV incidence reported to the National System of Public Health Surveillance in 27 municipalities of Colombia between January 1 2015 and December 31 2017. We demonstrate that our model can represent the infection patterns over this time period with high confidence. In addition, we argue that the degree of under-reporting is also well estimated. Using the model we assess potential viability of public health scenarios for mitigating disease spread and find that targeting the sexual pathway alone has negligible impact on overall spread, but if the proportion of risky sexual behavior increases then it may become important. Targeting mosquitoes remains the best approach of those considered. These results may be useful for public health organizations and governments to construct and implement suitable health policies and reduce the impact of the Zika outbreaks.
Collapse
|
50
|
Sher AA, Lao YT, Coombs KM. HLA-A, HSPA5, IGFBP5 and PSMA2 Are Restriction Factors for Zika Virus Growth in Astrocytic Cells. Viruses 2022; 15:97. [PMID: 36680137 PMCID: PMC9863221 DOI: 10.3390/v15010097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
(1) Background: Zika virus (ZIKV), an arbo-flavivirus, is transmitted via Aeges aegyptii mosquitoes Following its major outbreaks in 2013, 2014 and 2016, WHO declared it a Public Health Emergency of International Concern. Symptoms of ZIKV infection include acute fever, conjunctivitis, headache, muscle & joint pain and malaise. Cases of its transmission also have been reported via perinatal, sexual and transfusion transmission. ZIKV pathologies include meningo-encephalitis and myelitis in the central nervous system (CNS) and Guillain-Barré syndrome and acute transient polyneuritis in the peripheral nervous system (PNS). Drugs like azithromycin have been tested as inhibitors of ZIKV infection but no vaccines or treatments are currently available. Astrocytes are the most abundant cells in the CNS and among the first cells in CNS infected by ZIKV; (2) Methods: We previously used SOMAScan proteomics to study ZIKV-infected astrocytic cells. Here, we use mass spectrometric analyses to further explain dysregulations in the cellular expression profile of glioblastoma astrocytoma U251 cells. We also knocked down (KD) some of the U251 cellular proteins using siRNAs and observed the impact on ZIKV replication and infectivity; (3) Results & Conclusions: The top ZIKV dysregulated cellular networks were antimicrobial response, cell death, and energy production while top dysregulated functions were antigen presentation, viral replication and cytopathic impact. Th1 and interferon signaling pathways were among the top dysregulated canonical pathways. siRNA-mediated KD of HLA-A, IGFBP5, PSMA2 and HSPA5 increased ZIKV titers and protein synthesis, indicating they are ZIKV restriction factors. ZIKV infection also restored HLA-A expression in HLA-A KD cells by 48 h post-infection, suggesting interactions between this gene product and ZIKV.
Collapse
Affiliation(s)
- Affan A. Sher
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ying Tenny Lao
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Kevin M. Coombs
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|