51
|
Ahmad N, Badshah SL, Junaid M, Ur Rehman A, Muhammad A, Khan K. Structural insights into the Zika virus NS1 protein inhibition using a computational approach. J Biomol Struct Dyn 2020; 39:3004-3011. [PMID: 32321364 DOI: 10.1080/07391102.2020.1759453] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Zika virus is part of the flaviviruses that spread through the Aedes mosquito species and causes neurological infectious diseases. The non-structural protein 1 (NS1) is an essential enzyme that is involved in the replication of Zika virus. In this study, the newly isolated flavonoid analogs were docked against the NS1 protein. Most of the compounds showed strong interactions with favorable binding energies in the active site of NS1. One of the suitable docked ligand-protein complexes was simulated along with the apo form of the enzyme for 100 ns. The simulation results validated the docking data. The molecular dynamics simulation analysis comprising of root mean square deviation and fluctuation, the radius of gyration, hydrogen bonding, potential energy, principle component analysis, and MM/PBSA revealed about the stability of the apo and complex systems. These flavonoids analogs can inhibit the hexamerization of the NS1 which is necessary for the Zika virus replication.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Nasir Ahmad
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Syed Lal Badshah
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Muhammad Junaid
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ashfaq Ur Rehman
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Akhtar Muhammad
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Khalid Khan
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| |
Collapse
|
52
|
Winokur OC, Main BJ, Nicholson J, Barker CM. Impact of temperature on the extrinsic incubation period of Zika virus in Aedes aegypti. PLoS Negl Trop Dis 2020; 14:e0008047. [PMID: 32187187 PMCID: PMC7105136 DOI: 10.1371/journal.pntd.0008047] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 03/30/2020] [Accepted: 01/09/2020] [Indexed: 11/18/2022] Open
Abstract
Since Zika virus (ZIKV) emerged as a global human health threat, numerous studies have pointed to Aedes aegypti as the primary vector due to its high competence and propensity to feed on humans. The majority of vector competence studies have been conducted between 26-28°C, but arboviral extrinsic incubation periods (EIPs), and therefore transmission efficiency, are known to be affected strongly by temperature. To better understand the relationship between ZIKV EIPs and temperature, we evaluated the effect of adult mosquito exposure temperature on ZIKV infection, dissemination, and transmission in Ae. aegypti at four temperatures: 18°C, 21°C, 26°C, and 30°C. Mosquitoes were exposed to viremic mice infected with a 2015 Puerto Rican ZIKV strain, and engorged mosquitoes were sorted into the four temperatures with 80% RH and constant access to 10% sucrose. ZIKV infection, dissemination, and transmission rates were assessed via RT-qPCR from individual mosquito bodies, legs and wings, and saliva, respectively, at three to five time points per temperature from three to 31 days, based on expectations from other flavivirus EIPs. The median time from ZIKV ingestion to transmission (median EIP, EIP50) at each temperature was estimated by fitting a generalized linear mixed model for each temperature. EIP50 ranged from 5.1 days at 30°C to 24.2 days at 21°C. At 26°C, EIP50 was 9.6 days. At 18°C, only 15% transmitted by day 31 so EIP50 could not be estimated. This is among the first studies to characterize the effects of temperature on ZIKV EIP in Ae. aegypti, and the first to do so based on feeding of mosquitoes on a live, viremic host. This information is critical for modeling ZIKV transmission dynamics to understand geographic and seasonal limits of ZIKV risk; it is especially relevant for determining risk in subtropical regions with established Ae. aegypti populations and relatively high rates of return travel from the tropics (e.g. California or Florida), as these regions typically experience cooler temperature ranges than tropical regions.
Collapse
Affiliation(s)
- Olivia C. Winokur
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
- Graduate Group of Entomology, University of California, Davis, California, United States of America
| | - Bradley J. Main
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Jay Nicholson
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Christopher M. Barker
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| |
Collapse
|
53
|
Peçanha PM, Gomes Junior SC, Pone SM, Pone MVDS, Vasconcelos Z, Zin A, Vilibor RHH, Costa RP, Meio MDBB, Nielsen-Saines K, Brasil P, Brickley E, Lopes Moreira ME. Neurodevelopment of children exposed intra-uterus by Zika virus: A case series. PLoS One 2020; 15:e0229434. [PMID: 32109947 PMCID: PMC7048286 DOI: 10.1371/journal.pone.0229434] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 02/06/2020] [Indexed: 11/23/2022] Open
Abstract
The main goal of this manuscript was to investigate the neurodevelopment of children exposed by Zika virus in the intrauterine period who are asymptomatic at birth. Newborns with documented Zika virus exposure during the intrauterine period who were asymptomatic at birth were followed in the first two years of life for neurodevelopment using Bayley III test. Children were classified as having normal or delayed neurodevelopment for age based on most recent Bayley III evaluation results. Eighty-four infants were included in the study. The first Bayley III evaluation was performed at a mean chronological age of 9.7±3.1 month; 13 children (15%) had a delay in one of the three domains, distributed as follow: 10 (12%) in the language domain and 3 (3.5%) in the motor domain. The most recent Bayley III evaluation was performed at a mean age 15.3±3.1 months; 42 children (50%) had a delay in one of the three domains: 4 (5%) in cognition, 31 (37%) in language, and 20 (24%) in motor performance. There were no statistical differences in Gender, Gestational Age, Birth Weight and Head Circurference at birth between children with normal and delayed neurodevelopment for age. A very high proportion of children exposed ZIKV during pregnancy who were asymptomatic at birth demonstrated a delay in neurodevelopment, mainly in the language domain, the first two years of life.
Collapse
Affiliation(s)
| | | | | | | | | | - Andrea Zin
- Instituto Fernandes Figueira–Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | - Karin Nielsen-Saines
- Universidade da California- UCLA, Los Angeles, California, United States of America
| | - Patricia Brasil
- Instituto Nacional de Infectologia-Fiocruz, Rio de Janeiro, Brazil
| | - Elizabeth Brickley
- London School of Hygiene & Tropical Medicine, London, England, United Kingdom
| | | |
Collapse
|
54
|
Tryptophan Trimers and Tetramers Inhibit Dengue and Zika Virus Replication by Interfering with Viral Attachment Processes. Antimicrob Agents Chemother 2020; 64:AAC.02130-19. [PMID: 31932383 DOI: 10.1128/aac.02130-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/25/2019] [Indexed: 12/15/2022] Open
Abstract
Here, we report a class of tryptophan trimers and tetramers that inhibit (at low micromolar range) dengue and Zika virus infection in vitro These compounds (AL family) have three or four peripheral tryptophan moieties directly linked to a central scaffold through their amino groups; thus, their carboxylic acid groups are free and exposed to the periphery. Structure-activity relationship (SAR) studies demonstrated that the presence of extra phenyl rings with substituents other than COOH at the N1 or C2 position of the indole side chain is a requisite for the antiviral activity against both viruses. The molecules showed potent antiviral activity, with low cytotoxicity, when evaluated on different cell lines. Moreover, they were active against laboratory and clinical strains of all four serotypes of dengue virus as well as a selected group of Zika virus strains. Additional mechanistic studies performed with the two most potent compounds (AL439 and AL440) demonstrated an interaction with the viral envelope glycoprotein (domain III) of dengue 2 virus, preventing virus attachment to the host cell membrane. Since no antiviral agent is approved at the moment against these two flaviviruses, further pharmacokinetic studies with these molecules are needed for their development as future therapeutic/prophylactic drugs.
Collapse
|
55
|
In Vitro Zika Virus Infection of Human Neural Progenitor Cells: Meta-Analysis of RNA-Seq Assays. Microorganisms 2020; 8:microorganisms8020270. [PMID: 32079323 PMCID: PMC7074932 DOI: 10.3390/microorganisms8020270] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/20/2022] Open
Abstract
The Zika virus (ZIKV) is an emergent arthropod-borne virus (arbovirus) responsible for congenital Zika syndrome (CZS) and a range of other congenital malformations. Evidence shows that ZIKV infects human neural progenitor cells (hNPCs) in the fetal brain, prompting inflammation and tissue damage/loss. Despite recent advances, little is known about the pathways involved in CZS pathogenesis. We performed a meta-analysis, gene ontology (GO), and pathway analysis of whole transcriptome studies with the aim of clarifying the genes and pathways potentially altered during hNPCs infection with ZIKV. We selected three studies (17 samples of infected hPNCs compared to hPNCs uninfected controls) through a systematic search of the Gene Expression Omnibus (GEO) database. The raw reads were trimmed, counted, and normalized. Next, we performed a rank product meta-analysis to detect consistently differentially expressed genes (DEGs) in these independent experiments. We detected 13 statistically significant DEGs. GO ontology and reactome analysis showed an enrichment of interferon, pro-inflammatory, and chemokines signaling and apoptosis pathways in ZIKV-infected cells. Moreover, we detected three possible new candidate genes involved in hNPCs infection: APOL6, XAF1, and TNFRSF1. Our results confirm that interferon (IFN) signaling dominates the ZIKV response, and that a crucial contribution is given by apoptotic pathways, which might elicit the CZS phenotype.
Collapse
|
56
|
Roldán JS, Cassola A, Castillo DS. Optimization of recombinant Zika virus NS1 protein secretion from HEK293 cells. ACTA ACUST UNITED AC 2020; 25:e00434. [PMID: 32095434 PMCID: PMC7033529 DOI: 10.1016/j.btre.2020.e00434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/09/2020] [Accepted: 02/10/2020] [Indexed: 12/17/2022]
Abstract
Stable recombinant ZIKV NS1-His-expressing HEK293 cells were generated. Rapamycin treatment followed by serum starvation leads to a 29-fold increase in recombinant ZIKV NS1 protein secretion. The purified recombinant ZIKV NS1 hexamer is a reliable biological tool for clinical diagnosis and surveillance purposes.
Sensitive, accurate and cost-effective diagnostic tests are urgently needed to detect Zika virus (ZIKV) infection. Nonstructural 1 (NS1) glycoprotein is an excellent diagnostic marker since it is released in a hexameric conformation from infected cells into the patient's bloodstream early in the course of the infection. We established a stable rZNS1-His-expression system in HEK293 cells through lentiviral transduction. A novel optimization approach to enhance rZNS1-His protein secretion in the mammalian expression system was accomplished through 50 nM rapamycin incubation followed by serum-free media incubation for 9 days, reaching protein yields of ∼10 mg/l of culture medium. Purified rZNS1-His hexamer was recognized by anti-NS1 antibodies in ZIKV patient's serum, and showed the ability to induce a humoral response in immunized mice. The obtained recombinant protein is a reliable biological tool that can potentially be applied in the development of diagnostic tests to detect ZIKV in infected patients during the acute phase.
Collapse
Affiliation(s)
- Julieta S Roldán
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde" (IIBIO), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Alejandro Cassola
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde" (IIBIO), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| | - Daniela S Castillo
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde" (IIBIO), Universidad Nacional de San Martín (UNSAM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Martín, Buenos Aires, Argentina
| |
Collapse
|
57
|
Cerebrospinal fluid immunoglobulins are increased in neonates exposed to Zika virus during foetal life. J Infect 2020; 80:419-425. [PMID: 31981639 DOI: 10.1016/j.jinf.2020.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/18/2019] [Accepted: 01/14/2020] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To compare immunoglobulin levels in cerebrospinal fluid (CSF) of neonates exposed to Zika virus (ZIKV) during foetal life (cases) with levels in CSF of control neonates. METHODS We identified 16 neonates who underwent lumbar puncture (LP), during the ZIKV epidemic (December/2015 to March/2016) whose mothers reported ZIKV clinical symptoms during gestation (cases). Congenital microcephaly was defined as head circumference ≤31.9 cm (boys) and ≤31.5 cm (girls) for term neonates, or ≤2 standard deviations below the mean for premature (<37 weeks) neonates. Subsequently, we identified neonates who underwent LP in the same lab and fulfilled criteria to be controls: age ≤4 days, CSF white blood cell count ≤8/mm3, CSF protein ≤132 mg/dL, CSF red blood cell count ≤1,000/mm3, neither central nervous system illness, nor congenital infection, nor microcephaly. CSF immunoglobulin concentrations were measured by mass spectrometry. RESULTS 13 controls were included. IgM, IgA, IgG, IgK, and IgL were significantly higher among cases (p < 0.001). Eight (50%) ZIKV exposed infants had congenital microcephaly. These showed the strongest immunoglobulin elevation of the IgM and IgA classes. CONCLUSION Neonates exposed to ZIKV infection during gestation present with elevated distinct immunoglobulins in CSF, both in cases that developed microcephaly and in cases that did not.
Collapse
|
58
|
Silva S, Shimizu JF, Oliveira DMD, Assis LRD, Bittar C, Mottin M, Sousa BKDP, Mesquita NCDMR, Regasini LO, Rahal P, Oliva G, Perryman AL, Ekins S, Andrade CH, Goulart LR, Sabino-Silva R, Merits A, Harris M, Jardim ACG. A diarylamine derived from anthranilic acid inhibits ZIKV replication. Sci Rep 2019; 9:17703. [PMID: 31776405 PMCID: PMC6881388 DOI: 10.1038/s41598-019-54169-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 10/29/2019] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-transmitted Flavivirus, originally identified in Uganda in 1947 and recently associated with a large outbreak in South America. Despite extensive efforts there are currently no approved antiviral compounds for treatment of ZIKV infection. Here we describe the antiviral activity of diarylamines derived from anthranilic acid (FAMs) against ZIKV. A synthetic FAM (E3) demonstrated anti-ZIKV potential by reducing viral replication up to 86%. We analyzed the possible mechanisms of action of FAM E3 by evaluating the intercalation of this compound into the viral dsRNA and its interaction with the RNA polymerase of bacteriophage SP6. However, FAM E3 did not act by these mechanisms. In silico results predicted that FAM E3 might bind to the ZIKV NS3 helicase suggesting that this protein could be one possible target of this compound. To test this, the thermal stability and the ATPase activity of the ZIKV NS3 helicase domain (NS3Hel) were investigated in vitro and we demonstrated that FAM E3 could indeed bind to and stabilize NS3Hel.
Collapse
Affiliation(s)
- Suely Silva
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Jacqueline Farinha Shimizu
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Débora Moraes de Oliveira
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil
| | - Leticia Ribeiro de Assis
- Laboratory of Antibiotics and Chemotherapeutics, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Cintia Bittar
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Melina Mottin
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | - Bruna Katiele de Paula Sousa
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | | | - Luis Octávio Regasini
- Laboratory of Antibiotics and Chemotherapeutics, São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Paula Rahal
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil
| | - Glaucius Oliva
- Institute of Physics of São Carlos, University of São Paulo, São Carlos, Brazil
| | - Alexander Luke Perryman
- Department of Pharmacology, Physiology and Neuroscience, Rutgers University-New Jersey Medical School, Newark, NJ 07103, United States
- Repare Therapeutics, 7210 Rue Frederick-Banting, Suite 100, Montreal, QC, H4S 2A1, Canada
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC, 27606, United States
| | - Carolina Horta Andrade
- LabMol - Laboratory of Molecular Modeling and Drug Design, Faculdade de Farmácia, Universidade Federal de Goias, Goiânia, GO, 74605- 170, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Federal University of Uberlandia, Uberlândia, MG, Brasil
| | - Robinson Sabino-Silva
- Integrative Physiology and Salivary Nanobiotechnology Group, Federal University of Uberlandia, Uberlândia, MG, Brasil
| | - Andres Merits
- Institute of Technology, University of Tartu, Nooruse 1, 50411, Tartu, Estonia
| | - Mark Harris
- School of Molecular and Cellular Biology, Faculty of Biological Sciences and Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlandia, Uberlândia, MG, Brazil.
- São Paulo State University, IBILCE, S. José do Rio Preto, SP, Brazil.
| |
Collapse
|
59
|
Millies B, von Hammerstein F, Gellert A, Hammerschmidt S, Barthels F, Göppel U, Immerheiser M, Elgner F, Jung N, Basic M, Kersten C, Kiefer W, Bodem J, Hildt E, Windbergs M, Hellmich UA, Schirmeister T. Proline-Based Allosteric Inhibitors of Zika and Dengue Virus NS2B/NS3 Proteases. J Med Chem 2019; 62:11359-11382. [DOI: 10.1021/acs.jmedchem.9b01697] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Benedikt Millies
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Franziska von Hammerstein
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Andrea Gellert
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Stefan Hammerschmidt
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Fabian Barthels
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Ulrike Göppel
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Melissa Immerheiser
- Institut für Virologie und Immunbiologie, Julius-Maximilians-Universität Würzburg, Versbacher Straße 7, 97078 Würzburg, Germany
| | - Fabian Elgner
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Nathalie Jung
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Michael Basic
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Christian Kersten
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Werner Kiefer
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| | - Jochen Bodem
- Institut für Virologie und Immunbiologie, Julius-Maximilians-Universität Würzburg, Versbacher Straße 7, 97078 Würzburg, Germany
| | - Eberhard Hildt
- Department of Virology, Paul-Ehrlich-Institut, 63225 Langen, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Straße 15, 60438 Frankfurt am Main, Germany
| | - Ute A. Hellmich
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Johann-Joachim-Becherweg 30, 55128 Mainz, Germany
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe-Universität, 60438 Frankfurt am Main, Germany
| | - Tanja Schirmeister
- Institute of Pharmacy and Biochemistry, Johannes Gutenberg University Mainz, Staudinger Weg 5, 55128 Mainz, Germany
| |
Collapse
|
60
|
Sharma V, Sharma M, Dhull D, Sharma Y, Kaushik S, Kaushik S. Zika virus: an emerging challenge to public health worldwide. Can J Microbiol 2019; 66:87-98. [PMID: 31682478 DOI: 10.1139/cjm-2019-0331] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Zika virus (ZIKV) is a mosquito-borne virus that was first isolated from Zika forest, Uganda, in 1947. Since its inception, major and minor outbreaks have been documented from several parts of world. Aedes spp. mosquitoes are the primary vectors of ZIKV, but the virus can also be transmitted through sexual practices, materno-fetal transmission, and blood transfusion. The clinical presentations of symptomatic ZIKV infections are similar to dengue and chikungunya, including fever, headache, arthralgia, retro-orbital pain, conjunctivitis, and rash. ZIKV often causes mild illness in the majority of cases, but in some instances, it is linked with congenital microcephaly and autoimmune disorders like Guillain-Barré syndrome. The recent Indian ZIKV outbreak suggests that the virus is circulating in the South East Asian region and may cause new outbreaks in future. At present, no specific vaccines or antivirals are available to treat ZIKV, so management and control of ZIKV infections rely mostly on preventive measures.
Collapse
Affiliation(s)
- Vikrant Sharma
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Manisha Sharma
- Department of Biotechnology, Himachal Pradesh University, Shimla-171005, Himachal Pradesh, India
| | - Divya Dhull
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Yashika Sharma
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Sulochana Kaushik
- Department of Genetics, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| | - Samander Kaushik
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak-124001, Haryana, India
| |
Collapse
|
61
|
Thompson EL, Vamos CA, Jones J, Liggett LG, Griner SB, G Logan R, Daley EM. Perceptions of Zika Virus Prevention Among College Students in Florida. J Community Health 2019; 43:673-679. [PMID: 29380211 DOI: 10.1007/s10900-018-0468-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Zika virus in Florida prompted a strong public health response, due to its causal association with birth defects. While primarily spread by mosquitos, Zika can be transmitted sexually. The spread of Zika may influence reproductive behaviors among sexually active persons in Florida. This study examined factors associated with willingness to change birth control method use in response to Zika virus among college women and men in Florida. Women and men ages 18-44 at a Florida university (N = 328) were surveyed about Zika knowledge, beliefs about Zika, use of contraceptives and condoms, and socio-demographics between November 2016-April 2017. The outcome variable was willingness to change birth control method were Zika in their area. Logistic regression models in SAS 9.4 were used. Most participants were women (80%), and 47% were 20-22 years old. Only 27% of participants said they would change their birth control method if Zika were in their area. Participants who knew that Zika was sexually transmitted were more likely to be willing to change their birth control method (aOR = 1.71, 95%CI 1.01-2.91). Participants who agreed or strongly agreed that they were fearful of being infected with Zika virus were more likely to be willing to change their birth control methods (aOR = 1.98, 95%CI 1.07-3.67). This study found that, among Florida college students, Zika beliefs and knowledge were associated with a willingness to change birth control method in response to Zika. Understanding the factors that motivate individuals to change reproductive behaviors during an emerging health issue can help tailor preventative messages.
Collapse
Affiliation(s)
- Erika L Thompson
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA. .,Department of Health Behavior and Health Systems, School of Public Health, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| | - Cheryl A Vamos
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| | - Julianna Jones
- Department of Global Health and Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| | - Langdon G Liggett
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| | - Stacey B Griner
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| | - Rachel G Logan
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| | - Ellen M Daley
- Department of Community and Family Health, College of Public Health, University of South Florida, 13201 Bruce B Downs Blvd, MDC 56, Tampa, FL, 33612, USA
| |
Collapse
|
62
|
Guo Q, Chan JFW, Poon VKM, Wu S, Chan CCS, Hou L, Yip CCY, Ren C, Cai JP, Zhao M, Zhang AJ, Song X, Chan KH, Wang B, Kok KH, Wen Y, Yuen KY, Chen W. Immunization With a Novel Human Type 5 Adenovirus-Vectored Vaccine Expressing the Premembrane and Envelope Proteins of Zika Virus Provides Consistent and Sterilizing Protection in Multiple Immunocompetent and Immunocompromised Animal Models. J Infect Dis 2019; 218:365-377. [PMID: 29617816 DOI: 10.1093/infdis/jiy187] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/28/2018] [Indexed: 12/30/2022] Open
Abstract
Background Zika virus (ZIKV) infection may be associated with severe complications and disseminated via both vector-borne and nonvector-borne routes. Adenovirus-vectored vaccines represent a favorable controlling measure for the ZIKV epidemic because they have been shown to be safe, immunogenic, and rapidly generable for other emerging viral infections. Evaluations of 2 previously reported adenovirus-vectored ZIKV vaccines were performed using nonlethal animal models and/or nonepidemic ZIKV strain. Methods We constructed 2 novel human adenovirus 5 (Ad5)-vectored vaccines containing the ZIKV premembrane-envelope (Ad5-Sig-prM-Env) and envelope (Ad5-Env) proteins, respectively, and evaluated them in multiple nonlethal and lethal animal models using epidemic ZIKV strains. Results Both vaccines elicited robust humoral and cellular immune responses in immunocompetent BALB/c mice. Dexamethasone-immunosuppressed mice vaccinated with either vaccine demonstrated robust and durable antibody responses and significantly lower blood and tissue viral loads than controls (P < .05). Similar findings were also observed in interferon-α/β receptor-deficient A129 mice. In both of these immunocompromised animal models, Ad5-Sig-prM-Env-vaccinated mice had significantly (P < .05) higher titers of anti-ZIKV-specific neutralizing antibody titers and lower (undetectable) viral loads than Ad5-Env-vaccinated mice. The close correlation between the neutralizing antibody titer and viral load helped to explain the better protective effect of Ad5-Sig-prM-Env than Ad5-Env. Anamnestic response was absent in Ad5-Sig-prM-Env-vaccinated A129 mice. Conclusions Ad5-Sig-prM-Env provided sterilizing protection against ZIKV infection in mice.
Collapse
Affiliation(s)
- Qiang Guo
- Beijing Institute of Biotechnology, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Vincent Kwok-Man Poon
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Shipo Wu
- Beijing Institute of Biotechnology, China
| | - Chris Chung-Sing Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lihua Hou
- Beijing Institute of Biotechnology, China
| | - Cyril Chik-Yan Yip
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Anna Jinxia Zhang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | | | - Kwok-Hung Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Busen Wang
- Beijing Institute of Biotechnology, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yanbo Wen
- Beijing Institute of Biotechnology, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,Carol Yu Centre for Infection, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China.,The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wei Chen
- Beijing Institute of Biotechnology, China
| |
Collapse
|
63
|
Kuper H, Lyra TM, Moreira MEL, de Albuquerque MDSV, de Araújo TVB, Fernandes S, Jofre-Bonet M, Larson H, Lopes de Melo AP, Mendes CHF, Moreira MCN, do Nascimento MAF, Penn-Kekana L, Pimentel C, Pinto M, Simas C, Valongueiro S. Social and economic impacts of congenital Zika syndrome in Brazil: Study protocol and rationale for a mixed-methods study. Wellcome Open Res 2019; 3:127. [PMID: 31667356 PMCID: PMC6807146 DOI: 10.12688/wellcomeopenres.14838.2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2019] [Indexed: 12/22/2022] Open
Abstract
Global concern broke out in late 2015 as thousands of children in Brazil were born with microcephaly, which was quickly linked to congenital infection with Zika virus (ZIKV). ZIKV is now known to cause a wider spectrum of severe adverse outcomes-congenital Zika syndrome (CZS)-and also milder impairments. This study aimed to explore the social and economic impacts of CZS in Brazil. Data was collected through mixed methods across two settings: Recife City and Jaboatão dos Guararapes in Pernambuco State (the epicentre of the epidemic), and the city of Rio de Janeiro (where reports of ZIKV infection and CZS were less frequent). Data was collected May 2017-January 2018. Ethical standards were adhered to throughout the research. In-depth qualitative interviews were conducted with: mothers and other carers of children with CZS (approximately 30 per setting), pregnant women (10-12 per setting), men and women of child-bearing age (16-20 per setting), and health professionals (10-12 per setting). Thematic analysis was undertaken independently by researchers from at least two research settings, and these were shared for feedback. A case-control study was undertaken to quantitatively explore social and economic differences between caregivers of a child with CZS (cases) and caregivers with an unaffected child (controls). We aimed to recruit 100 cases and 100 controls per setting, from existing studies. The primary caregiver, usually the mother, was interviewed using a structured questionnaire to collect information on: depression, anxiety, stress, social support, family quality of life, health care and social service use, and costs incurred by families. Multivariable logistic regression analyses were used to compare outcomes for cases and controls. Costs incurred as a result of CZS were estimated from the perspective of the health system, families and society. Modelling was undertaken to estimate the total economic burden of CZS from those three perspectives.
Collapse
Affiliation(s)
- Hannah Kuper
- International Centre for Evidence in Disability, Clinical Research Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Tereza Maciel Lyra
- Aggeu Magalhães Institute, FIOCRUZ/PE, Recife, Brazil.,Faculty of Medicine, University of Pernambuco, Recife, Brazil
| | | | | | | | - Silke Fernandes
- Faculty of Public Health and Policy, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Heidi Larson
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Ana Paula Lopes de Melo
- Aggeu Magalhães Institute, FIOCRUZ/PE, Recife, Brazil.,Public Health Department, Federal University of Pernambuco, Recife, Brazil
| | | | | | | | - Loveday Penn-Kekana
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Marcia Pinto
- Fernando Figueira Maternal and Children's Institute, Rio de Janeiro, Brazil
| | - Clarissa Simas
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Sandra Valongueiro
- Postgraduate Programme in Public Health, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
64
|
Kuper H, Lyra TM, Moreira MEL, de Albuquerque MDSV, de Araújo TVB, Fernandes S, Jofre-Bonet M, Larson H, Lopes de Melo AP, Mendes CHF, Moreira MCN, do Nascimento MAF, Penn-Kekana L, Pimentel C, Pinto M, Simas C, Valongueiro S. Social and economic impacts of congenital Zika syndrome in Brazil: Study protocol and rationale for a mixed-methods study. Wellcome Open Res 2019; 3:127. [PMID: 31667356 DOI: 10.12688/wellcomeopenres.14838.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2018] [Indexed: 11/20/2022] Open
Abstract
Global concern broke out in late 2015 as thousands of children in Brazil were born with microcephaly, which was quickly linked to congenital infection with Zika virus (ZIKV). ZIKV is now known to cause a wider spectrum of severe adverse outcomes-congenital Zika syndrome (CZS)-and also milder impairments. This study aimed to explore the social and economic impacts of CZS in Brazil. Data was collected through mixed methods across two settings: Recife City and Jaboatão dos Guararapes in Pernambuco State (the epicentre of the epidemic), and the city of Rio de Janeiro (where reports of ZIKV infection and CZS were less frequent). Data was collected May 2017-January 2018. Ethical standards were adhered to throughout the research. In-depth qualitative interviews were conducted with: mothers and other carers of children with CZS (approximately 30 per setting), pregnant women (10-12 per setting), men and women of child-bearing age (16-20 per setting), and health professionals (10-12 per setting). Thematic analysis was undertaken independently by researchers from at least two research settings, and these were shared for feedback. A case-control study was undertaken to quantitatively explore social and economic differences between caregivers of a child with CZS (cases) and caregivers with an unaffected child (controls). We aimed to recruit 100 cases and 100 controls per setting, from existing studies. The primary caregiver, usually the mother, was interviewed using a structured questionnaire to collect information on: depression, anxiety, stress, social support, family quality of life, health care and social service use, and costs incurred by families. Multivariable logistic regression analyses were used to compare outcomes for cases and controls. Costs incurred as a result of CZS were estimated from the perspective of the health system, families and society. Modelling was undertaken to estimate the total economic burden of CZS from those three perspectives.
Collapse
Affiliation(s)
- Hannah Kuper
- International Centre for Evidence in Disability, Clinical Research Department, London School of Hygiene & Tropical Medicine, London, UK
| | - Tereza Maciel Lyra
- Aggeu Magalhães Institute, FIOCRUZ/PE, Recife, Brazil.,Faculty of Medicine, University of Pernambuco, Recife, Brazil
| | | | | | | | - Silke Fernandes
- Faculty of Public Health and Policy, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Heidi Larson
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Ana Paula Lopes de Melo
- Aggeu Magalhães Institute, FIOCRUZ/PE, Recife, Brazil.,Public Health Department, Federal University of Pernambuco, Recife, Brazil
| | | | | | | | - Loveday Penn-Kekana
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Marcia Pinto
- Fernando Figueira Maternal and Children's Institute, Rio de Janeiro, Brazil
| | - Clarissa Simas
- Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Sandra Valongueiro
- Postgraduate Programme in Public Health, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
65
|
Wilder-Smith A, Wei Y, de Araújo TVB, VanKerkhove M, Turchi Martelli CM, Turchi MD, Teixeira M, Tami A, Souza J, Sousa P, Soriano-Arandes A, Soria-Segarra C, Sanchez Clemente N, Rosenberger KD, Reveiz L, Prata-Barbosa A, Pomar L, Pelá Rosado LE, Perez F, Passos SD, Nogueira M, Noel TP, Moura da Silva A, Moreira ME, Morales I, Miranda Montoya MC, Miranda-Filho DDB, Maxwell L, Macpherson CNL, Low N, Lan Z, LaBeaud AD, Koopmans M, Kim C, João E, Jaenisch T, Hofer CB, Gustafson P, Gérardin P, Ganz JS, Dias ACF, Elias V, Duarte G, Debray TPA, Cafferata ML, Buekens P, Broutet N, Brickley EB, Brasil P, Brant F, Bethencourt S, Benedetti A, Avelino-Silva VL, Ximenes RADA, Alves da Cunha A, Alger J, Zika Virus Individual Participant Data Consortium
Abreu de carvalhoLiège MariaBatistaRosangelaBertozziAna PaulaCarlesGabrielCotrimDeniseDamascenoLuanaDimitrakisLadyDuarte rodriguesMaría ManoelaEstofoleteCassia FFragoso da silveira gouvêaMaria IsabelFumadó-pérezVickyGazetaRosa EstelaKaydos-danielsNeelyGilboaSuzanneKrystosikAmyLambertVéroniqueLópez-hortelanoMilagros GarcíaMussi-pinhataMarisa MarciaNelsonChristinaNielsenKarinOlianiDenise MRabelloRenataRibeiroMarizeliaRockxBarryRodriguesLaura CSalgadoSilviaSilveiraKatiaSulleiroElenaTongVanValenciaDianaDe souzaWayner VieiraVillar centenoLuis AngelZinAndrea. Understanding the relation between Zika virus infection during pregnancy and adverse fetal, infant and child outcomes: a protocol for a systematic review and individual participant data meta-analysis of longitudinal studies of pregnant women and their infants and children. BMJ Open 2019; 9:e026092. [PMID: 31217315 PMCID: PMC6588966 DOI: 10.1136/bmjopen-2018-026092] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 02/11/2019] [Accepted: 05/09/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Zika virus (ZIKV) infection during pregnancy is a known cause of microcephaly and other congenital and developmental anomalies. In the absence of a ZIKV vaccine or prophylactics, principal investigators (PIs) and international leaders in ZIKV research have formed the ZIKV Individual Participant Data (IPD) Consortium to identify, collect and synthesise IPD from longitudinal studies of pregnant women that measure ZIKV infection during pregnancy and fetal, infant or child outcomes. METHODS AND ANALYSIS We will identify eligible studies through the ZIKV IPD Consortium membership and a systematic review and invite study PIs to participate in the IPD meta-analysis (IPD-MA). We will use the combined dataset to estimate the relative and absolute risk of congenital Zika syndrome (CZS), including microcephaly and late symptomatic congenital infections; identify and explore sources of heterogeneity in those estimates and develop and validate a risk prediction model to identify the pregnancies at the highest risk of CZS or adverse developmental outcomes. The variable accuracy of diagnostic assays and differences in exposure and outcome definitions means that included studies will have a higher level of systematic variability, a component of measurement error, than an IPD-MA of studies of an established pathogen. We will use expert testimony, existing internal and external diagnostic accuracy validation studies and laboratory external quality assessments to inform the distribution of measurement error in our models. We will apply both Bayesian and frequentist methods to directly account for these and other sources of uncertainty. ETHICS AND DISSEMINATION The IPD-MA was deemed exempt from ethical review. We will convene a group of patient advocates to evaluate the ethical implications and utility of the risk stratification tool. Findings from these analyses will be shared via national and international conferences and through publication in open access, peer-reviewed journals. TRIAL REGISTRATION NUMBER PROSPERO International prospective register of systematic reviews (CRD42017068915).
Collapse
Affiliation(s)
- Annelies Wilder-Smith
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Yinghui Wei
- Centre for Mathematical Sciences, University of Plymouth, Plymouth, UK
| | | | - Maria VanKerkhove
- Health Emergencies Programme, Organisation mondiale de la Sante, Geneve, Switzerland
| | | | - Marília Dalva Turchi
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goiânia, Brazil
| | - Mauro Teixeira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adriana Tami
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, The Netherlands
| | - João Souza
- Department of Social Medicine, University of São Paulo, São Paulo, Brazil
| | - Patricia Sousa
- Reference Center for Neurodevelopment, Assistance, and Rehabilitation of Children, State Department of Health of Maranhão, Sao Luís, Brazil
| | | | | | | | - Kerstin Daniela Rosenberger
- Department of Infectious Diseases, Section Clinical Tropical Medicine, UniversitatsKlinikum Heidelberg, Heidelberg, Germany
| | - Ludovic Reveiz
- Evidence and Intelligence for Action in Health, Pan American Health Organization, Washington, District of Columbia, USA
| | - Arnaldo Prata-Barbosa
- Department of Pediatrics, D’Or Institute for Research & Education, Rio de Janeiro, Brazil
| | - Léo Pomar
- Department of Obstetrics and Gynecology, Centre Hospitalier de l’Ouest Guyanais, Saint-Laurent du Maroni, French Guiana
| | | | - Freddy Perez
- Communicable Diseases and Environmental Determinants of Health Department, Pan American Health Organization, Washington, District of Columbia, USA
| | | | - Mauricio Nogueira
- Faculdade de Medicina de Sao Jose do Rio Preto, Department of Dermatologic Diseases, São José do Rio Preto, Brazil
| | - Trevor P. Noel
- Windward Islands Research and Education Foundation, St. George’s University, True Blue Point, Grenada
| | - Antônio Moura da Silva
- Department of Public Health, Universidade Federal do Maranhão – São Luís, São Luís, Brazil
| | | | - Ivonne Morales
- Department of Infectious Diseases, Section Clinical Tropical Medicine, UniversitatsKlinikum Heidelberg, Heidelberg, Germany
| | | | | | - Lauren Maxwell
- Reproductive Health and Research, World Health Organization, Geneva, Switzerland
- Hubert Department of Global Health, Emory University, Atlanta, Georgia, USA
| | - Calum N. L. Macpherson
- Windward Islands Research and Education Foundation, St. George’s University, True Blue Point, Grenada
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Zhiyi Lan
- McGill University Health Centre, McGill University, Montréal, Canada
| | | | - Marion Koopmans
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caron Kim
- Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Esaú João
- Department of Infectious Diseases, Hospital Federal dos Servidores do Estado, Rio de Janeiro, Brazil
| | - Thomas Jaenisch
- Department of Infectious Diseases, Section Clinical Tropical Medicine, UniversitatsKlinikum Heidelberg, Heidelberg, Germany
| | - Cristina Barroso Hofer
- Instituto de Puericultura e Pediatria Martagão Gesteira, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paul Gustafson
- Statistics, University of British Columbia, British Columbia, Vancouver, Canada
| | - Patrick Gérardin
- INSERM CIC1410 Clinical Epidemiology, CHU La Réunion, Saint Pierre, Réunion
- UM 134 PIMIT (CNRS 9192, INSERM U1187, IRD 249, Université de la Réunion), Universite de la Reunion, Sainte Clotilde, Réunion
| | | | - Ana Carolina Fialho Dias
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vanessa Elias
- Sustainable Development and Environmental Health, Pan American Health Organization, Washington, District of Columbia, USA
| | - Geraldo Duarte
- Department of Gynecology and Obstetrics, University of São Paulo, São Paulo, Brazil
| | - Thomas Paul Alfons Debray
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - María Luisa Cafferata
- Mother and Children Health Research Department, Instituto de Efectividad Clinica y Sanitaria, Buenos Aires, Argentina
| | - Pierre Buekens
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, USA
| | - Nathalie Broutet
- Department of Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Elizabeth B. Brickley
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Patrícia Brasil
- Instituto de pesquisa Clínica Evandro Chagas, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Fátima Brant
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sarah Bethencourt
- Facultad de Ciencias de la Salud, Universidad de Carabobo, Valencia, Carabobo, Bolivarian Republic of Venezuela
| | - Andrea Benedetti
- Departments of Medicine and of Epidemiology, Biostatistics & Occupational Health, McGill University, Montreal, Quebec, Canada
| | - Vivian Lida Avelino-Silva
- Department of Infectious and Parasitic Diseases, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo, Brazil
| | | | | | - Jackeline Alger
- Facultad de Ciencias Médicas, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | | |
Collapse
|
66
|
Xu D, Li C, Qin CF, Xu Z. Update on the Animal Models and Underlying Mechanisms for ZIKV-Induced Microcephaly. Annu Rev Virol 2019; 6:459-479. [PMID: 31206355 DOI: 10.1146/annurev-virology-092818-015740] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The circulation of Zika virus (ZIKV) in nearly 80 countries and territories poses a significant global threat to public health. ZIKV is causally linked to severe developmental defects in the brain, recognized as congenital Zika syndrome (CZS), which includes microcephaly and other serious congenital neurological complications. Since the World Health Organization declared the ZIKV outbreak a public health emergency of international concern, remarkable progress has been made in the generation of different ZIKV infection animal models to gain insight into cellular targets and pathogenesis and to explore the associated underlying mechanisms. Here we focus on summarizing our current understanding of the effects of ZIKV on mammalian brain development in different developmental stages and discuss the potential underlying mechanisms of ZIKV-induced CZS, as well as future perspectives.
Collapse
Affiliation(s)
- Dan Xu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China;
| | - Cui Li
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; .,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
67
|
Zika Virus Subverts Stress Granules To Promote and Restrict Viral Gene Expression. J Virol 2019; 93:JVI.00520-19. [PMID: 30944179 PMCID: PMC6613768 DOI: 10.1128/jvi.00520-19] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Many viruses inhibit SGs. In this study, we observed that ZIKV restricts SG assembly, likely by relocalizing and subverting specific SG proteins to modulate ZIKV replication. This ZIKV-SG protein interaction is interesting, as many SG proteins are also known to function in neuronal granules, which are critical in neural development and function. Moreover, dysregulation of different SG proteins in neurons has been shown to play a role in the progression of neurodegenerative diseases. The likely consequences of ZIKV modulating SG assembly and subverting specific SG proteins are alterations to cellular mRNA transcription, splicing, stability, and translation. Such changes in cellular ribostasis could profoundly affect neural development and contribute to the devastating developmental and neurological anomalies observed following intrauterine ZIKV infection. Our study provides new insights into virus-host interactions and the identification of the SG proteins that may contribute to the unusual pathogenesis associated with this reemerging arbovirus. Flaviviruses limit the cell stress response by preventing the formation of stress granules (SGs) and modulate viral gene expression by subverting different proteins involved in the stress granule pathway. In this study, we investigated the formation of stress granules during Zika virus (ZIKV) infection and the role stress granule proteins play during the viral life cycle. Using immunofluorescence and confocal microscopy, we determined that ZIKV disrupted the formation of arsenite-induced stress granules and changed the subcellular distribution, but not the abundance or integrity, of stress granule proteins. We also investigated the role of different stress granule proteins in ZIKV infection by using target-specific short interfering RNAs to deplete Ataxin2, G3BP1, HuR, TIA-1, TIAR, and YB1. Knockdown of TIA-1 and TIAR affected ZIKV protein and RNA levels but not viral titers. Conversely, depletion of Ataxin2 and YB1 decreased virion production despite having only a small effect on ZIKV protein expression. Notably, however, depletion of G3BP1 and HuR decreased and increased ZIKV gene expression and virion production, respectively. Using an MR766 Gaussia Luciferase reporter genome together with knockdown and overexpression assays, G3BP1 and HuR were found to modulate ZIKV replication. These data indicate that ZIKV disrupts the formation of stress granules by sequestering stress granule proteins required for replication, where G3BP1 functions to promote ZIKV infection while HuR exhibits an antiviral effect. The results of ZIKV relocalizing and subverting select stress granule proteins might have broader consequences on cellular RNA homeostasis and contribute to cellular gene dysregulation and ZIKV pathogenesis. IMPORTANCE Many viruses inhibit SGs. In this study, we observed that ZIKV restricts SG assembly, likely by relocalizing and subverting specific SG proteins to modulate ZIKV replication. This ZIKV-SG protein interaction is interesting, as many SG proteins are also known to function in neuronal granules, which are critical in neural development and function. Moreover, dysregulation of different SG proteins in neurons has been shown to play a role in the progression of neurodegenerative diseases. The likely consequences of ZIKV modulating SG assembly and subverting specific SG proteins are alterations to cellular mRNA transcription, splicing, stability, and translation. Such changes in cellular ribostasis could profoundly affect neural development and contribute to the devastating developmental and neurological anomalies observed following intrauterine ZIKV infection. Our study provides new insights into virus-host interactions and the identification of the SG proteins that may contribute to the unusual pathogenesis associated with this reemerging arbovirus.
Collapse
|
68
|
Badshah SL, Ahmad N, Ur Rehman A, Khan K, Ullah A, Alsayari A, Muhsinah AB, N Mabkhot Y. Molecular docking and simulation of Zika virus NS3 helicase. BMC Chem 2019; 13:67. [PMID: 31384814 PMCID: PMC6661806 DOI: 10.1186/s13065-019-0582-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 05/02/2019] [Indexed: 02/06/2023] Open
Abstract
The Zika virus (ZIKV) has gained attention for the last few years due to the congenital microcephaly and Guillain–Barre Syndrome that resulted in humans. The non-structural protein-3 (NS3) helicase of ZIKV play an important role in viral RNA replication. In this article, we performed hundred nanosecond molecular dynamics simulation and molecular docking of the NS3 helicase of ZIKV with 1,4-benzothiazine derivatives. The root mean square deviation (RMSD) analyses showed the stability of the NS3 helicase. The simulation showed that the flexible and rigid domains of the protein play a crucial role during the RNA replication process. All such domains with ligand binding pockets can be targeted for drug design. The molecular docking showed that the strong hydrogen bonding and arene-cation interactions are responsible for the binding between NS3 and 1,4-benzothiazine derivatives, which provides a new dimension for potent drug design for ZIKV.
Collapse
Affiliation(s)
- Syed Lal Badshah
- 1Department of Chemistry, Islamia College University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Nasir Ahmad
- 1Department of Chemistry, Islamia College University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Ashfaq Ur Rehman
- 2State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240 China
| | - Khalid Khan
- 1Department of Chemistry, Islamia College University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Asad Ullah
- 1Department of Chemistry, Islamia College University, Peshawar, Khyber Pakhtunkhwa Pakistan
| | - Abdulrhman Alsayari
- 3Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529 Saudi Arabia
| | - Abdullatif Bin Muhsinah
- 3Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, 62529 Saudi Arabia
| | - Yahia N Mabkhot
- 4Department of Pharmaceutical Chemistry, College of Pharamacy, King Khalid University, Abha, 61441 Saudi Arabia
| |
Collapse
|
69
|
Yuan S, Chan JFW, Ye ZW, Wen L, Tsang TGW, Cao J, Huang J, Chan CCY, Chik KKH, Choi GKY, Cai JP, Yin F, Chu H, Liang M, Jin DY, Yuen KY. Screening of an FDA-Approved Drug Library with a Two-Tier System Identifies an Entry Inhibitor of Severe Fever with Thrombocytopenia Syndrome Virus. Viruses 2019; 11:v11040385. [PMID: 31027241 PMCID: PMC6520937 DOI: 10.3390/v11040385] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022] Open
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne bunyavirus that causes severe disease in humans with case-fatality rates of up to 30%. There are currently very limited treatment options for SFTSV infection. We conducted a drug repurposing program by establishing a two-tier test system to rapidly screen a Food and Drug Administration- (FDA)-approved drug library for drug compounds with anti-SFTSV activity in vitro. We identified five drug compounds that inhibited SFTSV replication at low micromolar concentrations, including hexachlorophene, triclosan, regorafenib, eltrombopag, and broxyquinoline. Among them, hexachlorophene was the most potent with an IC50 of 1.3 ± 0.3 µM and a selectivity index of 18.7. Mechanistic studies suggested that hexachlorophene was a virus entry inhibitor, which impaired SFTSV entry into host cells by interfering with cell membrane fusion. Molecular docking analysis predicted that the binding of hexachlorophene with the hydrophobic pocket between domain I and domain III of the SFTSV Gc glycoprotein was highly stable. The novel antiviral activity and mechanism of hexachlorophene in this study would facilitate the use of hexachlorophene as a lead compound to develop more entry inhibitors with higher anti-SFTSV potency and lower toxicity.
Collapse
Affiliation(s)
- Shuofeng Yuan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Zi-Wei Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Lei Wen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Terance Gi-Wai Tsang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jianli Cao
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jingjing Huang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Chris Chun-Yiu Chan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kenn Ka-Heng Chik
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Garnet Kwan-Yue Choi
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Jian-Piao Cai
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Feifei Yin
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Pathogen Biology, Hainan Medical University, Haikou 571101, China.
- Key Laboratory of Translational Tropical Medicine, Hainan Medical University, Haikou 571101, China.
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Mifang Liang
- Key Laboratory for Medical Virology and National Institute for Viral Disease Control and Prevention, Chinese Centre for Disease Control and Prevention, Beijing 102206, China.
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong, China.
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, Hainan Medical University, Haikou 571101, China, and The University of Hong Kong, Pokfulam, Hong Kong, China.
- The Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
70
|
Garbin CAS, Teruel GP, Saliba TA, Moimaz SAS, Garbin AJÍ. Knowledge and attitude of women with high-risk pregnancy about the Zika virus transmission. CIENCIA & SAUDE COLETIVA 2019; 26:233-240. [PMID: 33533844 DOI: 10.1590/1413-81232020261.25752018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/15/2019] [Indexed: 11/22/2022] Open
Abstract
Objective was to analyze the knowledge and attitude of high risk pregnant women about zika. This is a cross-sectional study, quantitative, with a sample of 201 high risk women who perform prenatal the Ambulatory Medical Specialties. A self-administered instrument, was applied after consultation with the doctor. Inclusion criteria were the presence of pregnant women on the day of the interview and their consent to participate. Exclusion criteria were those who did not agree to participate and were not performing prenatal care during the study period. For the data analysis we used chi square and fisher exact, in software Epi info 7.1 and Bioestat 5.0. Of the pregnant women, 76% believed that their neighborhood was likely to be infected by the virus and used measures to control mosquito proliferation, such as not leaving standing water (n = 154). In relation to knowledge, there was an association between Zika and microcephaly (p≤ 0.0001) and the need for more information (p = 0.0439). To prevent infection, 76% took no action, there was an association between the need for knowledge about the subject and the actions taken to combat the virus (p = 0.0049). We conclude that pregnant women's knowledge and attitude about zika is failed.
Collapse
Affiliation(s)
- Clea Adas Saliba Garbin
- Programa de Pós-Graduação em Odontologia Preventiva e Social, Departamento de Odontologia Infantil e Social, Faculdade de Odontologia de Araçatuba, Universidade Estadual de São Paulo. R. José Bonifácio 1193, Vila Mendonça. 16015-050 Araçatuba SP Brasil.
| | - Gabriela Peres Teruel
- Programa de Pós-Graduação em Odontologia Preventiva e Social, Departamento de Odontologia Infantil e Social, Faculdade de Odontologia de Araçatuba, Universidade Estadual de São Paulo. R. José Bonifácio 1193, Vila Mendonça. 16015-050 Araçatuba SP Brasil.
| | - Tânia Adas Saliba
- Programa de Pós-Graduação em Odontologia Preventiva e Social, Departamento de Odontologia Infantil e Social, Faculdade de Odontologia de Araçatuba, Universidade Estadual de São Paulo. R. José Bonifácio 1193, Vila Mendonça. 16015-050 Araçatuba SP Brasil.
| | - Suzely Adas Saliba Moimaz
- Programa de Pós-Graduação em Odontologia Preventiva e Social, Departamento de Odontologia Infantil e Social, Faculdade de Odontologia de Araçatuba, Universidade Estadual de São Paulo. R. José Bonifácio 1193, Vila Mendonça. 16015-050 Araçatuba SP Brasil.
| | - Artênio José Ísper Garbin
- Programa de Pós-Graduação em Odontologia Preventiva e Social, Departamento de Odontologia Infantil e Social, Faculdade de Odontologia de Araçatuba, Universidade Estadual de São Paulo. R. José Bonifácio 1193, Vila Mendonça. 16015-050 Araçatuba SP Brasil.
| |
Collapse
|
71
|
Hugo LE, Stassen L, La J, Gosden E, Ekwudu O, Winterford C, Viennet E, Faddy HM, Devine GJ, Frentiu FD. Vector competence of Australian Aedes aegypti and Aedes albopictus for an epidemic strain of Zika virus. PLoS Negl Trop Dis 2019; 13:e0007281. [PMID: 30946747 PMCID: PMC6467424 DOI: 10.1371/journal.pntd.0007281] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 04/16/2019] [Accepted: 03/05/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Recent epidemics of Zika virus (ZIKV) in the Pacific and the Americas have highlighted its potential as an emerging pathogen of global importance. Both Aedes (Ae.) aegypti and Ae. albopictus are known to transmit ZIKV but variable vector competence has been observed between mosquito populations from different geographical regions and different virus strains. Since Australia remains at risk of ZIKV introduction, we evaluated the vector competence of local Ae. aegypti and Ae. albopictus for a Brazilian epidemic ZIKV strain. In addition, we evaluated the impact of daily temperature fluctuations around a mean of 28°C on ZIKV transmission and extrinsic incubation period. METHODOLOGY/PRINCIPAL FINDINGS Mosquitoes were orally challenged with a Brazilian ZIKV strain (8.8 log CCID50/ml) and maintained at either 28°C constant or fluctuating temperature conditions. At 3, 7 and 14 days post-infection (dpi), ZIKV RNA copies were quantified in mosquito bodies, as well as wings and legs, using qRT-PCR, while virus antigen in saliva (a proxy for transmission) was detected using a cell culture ELISA. Despite high body and disseminated infection rates in both vectors, the transmission rates of ZIKV in saliva of Ae. aegypti (50-60%) were significantly higher than in Ae. albopictus (10%) at 14 dpi. Both species supported a high viral load in bodies, with no significant differences between constant and fluctuating temperature conditions. However, a significant difference in viral load in wings and legs between species was observed, with higher titres in Ae. aegypti maintained at constant temperature conditions. For ZIKV transmission to occur in Ae. aegypti, a disseminated virus load threshold of 7.59 log10 copies had to be reached. CONCLUSIONS/SIGNIFICANCE Australian Ae. aegypti are better able to transmit a Brazilian ZIKV strain than Ae. albopictus. The results are in agreement with the global consensus that Ae. aegypti is the major vector of ZIKV.
Collapse
Affiliation(s)
- Leon E. Hugo
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Liesel Stassen
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jessica La
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences Queensland University of Technology, Brisbane, Queensland, Australia
| | - Edward Gosden
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences Queensland University of Technology, Brisbane, Queensland, Australia
| | - O’mezie Ekwudu
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences Queensland University of Technology, Brisbane, Queensland, Australia
| | - Clay Winterford
- QIMR Berghofer Histotechnology Facility, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Elvina Viennet
- Research and Development, Australian Red Cross Blood Service, Brisbane, Queensland, Australia
| | - Helen M. Faddy
- Research and Development, Australian Red Cross Blood Service, Brisbane, Queensland, Australia
| | - Gregor J. Devine
- Mosquito Control Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Francesca D. Frentiu
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences Queensland University of Technology, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
72
|
Karesh JW, Mazzoli RA, Heintz SK. Ocular Manifestations of Mosquito-Transmitted Diseases. Mil Med 2019; 183:450-458. [PMID: 29635625 DOI: 10.1093/milmed/usx183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 12/22/2017] [Indexed: 11/13/2022] Open
Abstract
Of the 3,548 known mosquito species, about 100 transmit human diseases. Mosquitoes are distributed globally throughout tropical and temperate regions where standing water sources are available for egg laying and the maturation of larva. Female mosquitoes require blood meals for egg production. This is the main pathway for disease transmission. Mosquitoes carry several pathogenic organisms responsible for significant ocular pathology and vision loss including West Nile, Rift Valley, chikungunya, dengue viruses, various encephalitis viruses, malarial parasites, Francisella tularensis, microfilarial parasites, including Dirofilaria, Wuchereria, and Brugia spp., and human botfly larvae. Health care providers may not be familiar with many of these mosquito-transmitted diseases or their associated ocular findings delaying diagnosis, treatment, and recovery of visual function. This article aims to provide an overview of the ocular manifestations associated with mosquito-transmitted diseases.
Collapse
Affiliation(s)
- James W Karesh
- The Vision Center of Excellence, Walter Reed National Military Medical Center, 8960 Brown Drive, Building 2, 1st Floor, Room 1403-A, Bethesda, MD 20889-5629
| | - Robert A Mazzoli
- The Vision Center of Excellence, Walter Reed National Military Medical Center, 8960 Brown Drive, Building 2, 1 st Floor, Room 1403-A, Bethesda, MD 20889-5629.,Madigan Army Medical Center, 9040 Jackson Avenue, Tacoma, WA 98431
| | - Shannon K Heintz
- The Vision Center of Excellence, Walter Reed National Military Medical Center, 8960 Brown Drive, Building 2, 1st Floor, Room 1403-A, Bethesda, MD 20889-5629
| |
Collapse
|
73
|
Kurscheidt FA, Mesquita CSS, Damke GMZF, Damke E, Carvalho ARBDA, Suehiro TT, Teixeira JJV, da Silva VRS, Souza RP, Consolaro MEL. Persistence and clinical relevance of Zika virus in the male genital tract. Nat Rev Urol 2019; 16:211-230. [PMID: 30696994 DOI: 10.1038/s41585-019-0149-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Zika virus (ZIKV) is a re-emerging mosquito-transmitted flavivirus associated with congenital abnormalities in newborns and with Guillain-Barré syndrome in adults. The virus can also be sexually transmitted and can persist in the male genital tract. Studies evaluating the kinetics of ZIKV in seminal shedding of men who have been infected, as well as in animal and cellular models of infection, have shown that, in addition to the testis and epididymis, the prostate and seminal vesicles could also be involved in persistent ZIKV infection. Additionally, some studies have reported that men infected with ZIKV can present with genitourinary symptoms such as haematospermia, prostatitis, painful ejaculation, penile discharge, and oligospermia; however, little is known about the effect of ZIKV on fertility. Understanding the mechanisms that underlie persistent ZIKV infections in men is crucial to developing guidelines, effective vaccines, and therapies.
Collapse
Affiliation(s)
- Fábio A Kurscheidt
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Cristiane S S Mesquita
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Gabrielle M Z F Damke
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Edilson Damke
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Analine R B de A Carvalho
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Tamy T Suehiro
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Jorge J V Teixeira
- Division of Epidemiology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Vânia R S da Silva
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Raquel P Souza
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil
| | - Marcia E L Consolaro
- Laboratory of Clinical Cytology and Semen Analysis, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, PR, Brazil.
| |
Collapse
|
74
|
Gurung S, Reuter N, Preno A, Dubaut J, Nadeau H, Hyatt K, Singleton K, Martin A, Parks WT, Papin JF, Myers DA. Zika virus infection at mid-gestation results in fetal cerebral cortical injury and fetal death in the olive baboon. PLoS Pathog 2019; 15:e1007507. [PMID: 30657788 PMCID: PMC6355048 DOI: 10.1371/journal.ppat.1007507] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 01/31/2019] [Accepted: 12/05/2018] [Indexed: 11/21/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy in humans is associated with an increased incidence of congenital anomalies including microcephaly as well as fetal death and miscarriage and collectively has been referred to as Congenital Zika Syndrome (CZS). Animal models for ZIKV infection in pregnancy have been developed including mice and non-human primates (NHPs). In macaques, fetal CZS outcomes from maternal ZIKV infection range from none to significant. In the present study we develop the olive baboon (Papio anubis), as a model for vertical transfer of ZIKV during pregnancy. Four mid-gestation, timed-pregnant baboons were inoculated with the French Polynesian ZIKV isolate (104 ffu). This study specifically focused on the acute phase of vertical transfer. Dams were terminated at 7 days post infection (dpi; n = 1), 14 dpi (n = 2) and 21 dpi (n = 1). All dams exhibited mild to moderate rash and conjunctivitis. Viremia peaked at 5–7 dpi with only one of three dams remaining mildly viremic at 14 dpi. An anti-ZIKV IgM response was observed by 14 dpi in all three dams studied to this stage, and two dams developed a neutralizing IgG response by either 14 dpi or 21 dpi, the latter included transfer of the IgG to the fetus (cord blood). A systemic inflammatory response (increased IL2, IL6, IL7, IL15, IL16) was observed in three of four dams. Vertical transfer of ZIKV to the placenta was observed in three pregnancies (n = 2 at 14 dpi and n = 1 at 21 dpi) and ZIKV was detected in fetal tissues in two pregnancies: one associated with fetal death at ~14 dpi, and the other in a viable fetus at 21 dpi. ZIKV RNA was detected in the fetal cerebral cortex and other tissues of both of these fetuses. In the fetus studied at 21 dpi with vertical transfer of virus to the CNS, the frontal cerebral cortex exhibited notable defects in radial glia, radial glial fibers, disorganized migration of immature neurons to the cortical layers, and signs of pathology in immature oligodendrocytes. In addition, indices of pronounced neuroinflammation were observed including astrogliosis, increased microglia and IL6 expression. Of interest, in one fetus examined at 14 dpi without detection of ZIKV RNA in brain and other fetal tissues, increased neuroinflammation (IL6 and microglia) was observed in the cortex. Although the placenta of the 14 dpi dam with fetal death showed considerable pathology, only minor pathology was noted in the other three placentas. ZIKV was detected immunohistochemically in two placentas (14 dpi) and one placenta at 21 dpi but not at 7 dpi. This is the first study to examine the early events of vertical transfer of ZIKV in a NHP infected at mid-gestation. The baboon thus represents an additional NHP as a model for ZIKV induced brain pathologies to contrast and compare to humans as well as other NHPs. Zika virus is endemic in the Americas, primarily spread through mosquitos and sexual contact. Zika virus infection during pregnancy in women is associated with a variety of fetal pathologies now referred to as Congenital Zika Syndrome (CZS), with the most severe pathology being fetal microcephaly. Developing model organisms that faithfully recreate Zika infection in humans is critical for future development of treatments and preventions. In our present study, we infected Olive baboons at mid-gestation with Zika virus and studied the acute period of viremia and transfer of Zika virus to the fetus during the first three weeks after infection to better understand the timing and mechanisms of transfer of ZIKV across the placenta, leading to CZS. We observed Zika virus transfer to fetuses resulting in fetal death in one pregnancy and in a second pregnancy, significant damage to the frontal cortex of the fetal brain at a critical period of neurodevelopment in primates. Thus, the baboon provides a promising new non-human primate model to further compare and contrast the consequences of Zika virus infection in pregnancy to humans and other non-human primates.
Collapse
Affiliation(s)
- Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Nicole Reuter
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Alisha Preno
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jamie Dubaut
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Hugh Nadeau
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Kimberly Hyatt
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Krista Singleton
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Ashley Martin
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - W. Tony Parks
- Department of Pathology, University of Toronto, Toronto, Ontario, Canada
| | - James F. Papin
- Division of Comparative Medicine, Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- * E-mail:
| |
Collapse
|
75
|
Targeting SUMO Modification of the Non-Structural Protein 5 of Zika Virus as a Host-Targeting Antiviral Strategy. Int J Mol Sci 2019; 20:ijms20020392. [PMID: 30658479 PMCID: PMC6359730 DOI: 10.3390/ijms20020392] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/14/2019] [Accepted: 01/15/2019] [Indexed: 01/28/2023] Open
Abstract
Post-translational modifications of host or viral proteins are key strategies exploited by viruses to support virus replication and counteract host immune response. SUMOylation is a post-translational modification process mediated by a family of ubiquitin-like proteins called small ubiquitin-like modifier (SUMO) proteins. Multiple sequence alignment of 78 representative flaviviruses showed that most (72/78, 92.3%) have a putative SUMO-interacting motif (SIM) at their non-structural 5 (NS5) protein’s N-terminal domain. The putative SIM was highly conserved among 414 pre-epidemic and epidemic Zika virus (ZIKV) strains, with all of them having a putative SIM core amino acid sequence of VIDL (327/414, 79.0%) or VVDL (87/414, 21.0%). Molecular docking predicted that the hydrophobic SIM core residues bind to the β2 strand of the SUMO-1 protein, and the acidic residues flanking the core strengthen the binding through interactions with the basic surface of the SUMO protein. The SUMO inhibitor 2-D08 significantly reduced replication of flaviviruses and protected cells against ZIKV-induced cytopathic effects in vitro. A SIM-mutated ZIKV NS5 failed to efficiently suppress type I interferon signaling. Overall, these findings may suggest SUMO modification of the viral NS5 protein to be an evolutionarily conserved post-translational modification process among flaviviruses to enhance virus replication and suppress host antiviral response.
Collapse
|
76
|
Hasan S, Saeed S, Panigrahi R, Choudhary P. Zika Virus: A Global Public Health Menace: A Comprehensive Update. J Int Soc Prev Community Dent 2019; 9:316-327. [PMID: 31516865 PMCID: PMC6714416 DOI: 10.4103/jispcd.jispcd_433_18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/12/2019] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is a RNA virus and belongs to genus Flavivirus and family Flaviviridae. The virus was first discovered from a febrile primate from the Zika forests of Uganda in 1947 and the first human case was documented in 1954. The nonspecific clinical manifestations of ZIKV pose diagnostic dilemmas and delays early and effective treatment. Dental professionals should have a thorough knowledge about the virus and should follow standard infection control measures as the virus has been demonstrated in various body secretions (including salivary secretions). The disease is managed by symptomatic and supportive care and no vaccine exist till date. Recent ZIKV outbreaks and increase association of microcephaly with congenital ZIKV and neurological complications (Guillain-Barré syndrome) has drawn global public health attention. The World Health Organization declared it a public health emergency of international concern in 2016. This review article provides a detailed overview on ZIKV; it is clinical and oral manifestations, diagnostic aids, differential diagnosis, preventive aspects, and management protocol.
Collapse
Affiliation(s)
- Shamimul Hasan
- Department of Oral Medicine and Radiology, Faculty of Dentistry, Jamia Millia Islamia, New Delhi, India,Address for correspondence: Dr. Shamimul Hasan, C/O Mohd Javed Khan, C-4, Duplex Quarters, New Sir Syed Nagar, Aligarh, Uttar Pradesh, India. E-mail:
| | - Shazina Saeed
- Amity Institute of Public Health, Amity University, Noida, Uttar Pradesh, India
| | - Rajat Panigrahi
- Department of Oral Medicine and Radiology, Institute of Dental Sciences, SOA University, Bhubaneswar, Odisha, India
| | - Priyadarshini Choudhary
- Department of Oral Medicine and Radiology, Kalinga Institute of Dental Sciences, KIIT University, Bhubaneswar, Odisha, India
| |
Collapse
|
77
|
Mohr EL. Modeling Zika Virus-Associated Birth Defects in Nonhuman Primates. J Pediatric Infect Dis Soc 2018; 7:S60-S66. [PMID: 30590626 PMCID: PMC8506225 DOI: 10.1093/jpids/piy120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/31/2018] [Indexed: 01/22/2023]
Abstract
In utero infection with Zika virus (ZIKV) during pregnancy can lead to the development of birth defects and postnatal deficits. A nonhuman primate (NHP) model of congenital ZIKV infection can help fill the gaps in knowledge where tissue studies are required to define viral pathogenesis and identify targets for therapeutic intervention. This model system has already identified critical features of ZIKV pathogenesis in congenital infection. Before translating these NHP studies to human clinical trials, we must understand the similarities and differences between human and NHP fetal immune system development, neural development, and infant assessment tools. Because of the overall similarity between fetal and infant development in humans and NHPs, this NHP model can complement human clinical trials by defining immune correlates of protection and evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Emma L Mohr
- Department of Pediatrics, University of Wisconsin-Madison
| |
Collapse
|
78
|
The Scorpion Venom Peptide Smp76 Inhibits Viral Infection by Regulating Type-I Interferon Response. Virol Sin 2018; 33:545-556. [PMID: 30569290 DOI: 10.1007/s12250-018-0068-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/07/2018] [Indexed: 12/25/2022] Open
Abstract
Dengue virus (DENV) and Zika virus (ZIKV) have spread throughout many countries in the developing world and infect millions of people every year, causing severe harm to human health and the economy. Unfortunately, there are few effective vaccines and therapies available against these viruses. Therefore, the discovery of new antiviral agents is critical. Herein, a scorpion venom peptide (Smp76) characterized from Scorpio maurus palmatus was successfully expressed and purified in Escherichia coli BL21(DE3). The recombinant Smp76 (rSmp76) was found to effectively inhibit DENV and ZIKV infections in a dose-dependent manner in both cultured cell lines and primary mouse macrophages. Interestingly, rSmp76 did not inactivate the viral particles directly but suppressed the established viral infection, similar to the effect of interferon (IFN)-β. Mechanistically, rSmp76 was revealed to upregulate the expression of IFN-β by activating interferon regulatory transcription factor 3 (IRF3) phosphorylation, enhancing the type-I IFN response and inhibiting viral infection. This mechanism is significantly different from traditional virucidal antimicrobial peptides (AMPs). Overall, the scorpion venom peptide Smp76 is a potential new antiviral agent with a unique mechanism involving type-I IFN responses, demonstrating that natural AMPs can enhance immunity by functioning as immunomodulators.
Collapse
|
79
|
Abstract
How virulence evolves after a virus jumps to a new host species is central to disease emergence. Our current understanding of virulence evolution is based on insights drawn from two perspectives that have developed largely independently: long-standing evolutionary theory based on limited real data examples that often lack a genomic basis, and experimental studies of virulence-determining mutations using cell culture or animal models. A more comprehensive understanding of virulence mutations and their evolution can be achieved by bridging the gap between these two research pathways through the phylogenomic analysis of virus genome sequence data as a guide to experimental study.
Collapse
Affiliation(s)
- Jemma L Geoghegan
- Department of Biological Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Edward C Holmes
- Marie Bashir Institute for Infectious Diseases and Biosecurity, Charles Perkins Centre, School of Life and Environmental Sciences and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
80
|
Mumtaz N, Koedam M, van den Doel PB, van Leeuwen JPTM, Koopmans MPG, van der Eerden BCJ, Rockx B. Zika virus infection perturbs osteoblast function. Sci Rep 2018; 8:16975. [PMID: 30451958 PMCID: PMC6242880 DOI: 10.1038/s41598-018-35422-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/31/2018] [Indexed: 01/08/2023] Open
Abstract
Zika virus (ZIKV) infection is typically characterized by a mild self-limiting disease presenting with fever, rash, myalgia and arthralgia and severe fetal complications during pregnancy such as microcephaly, subcortical calcifications and arthrogyropsis. Virus-induced arthralgia due to perturbed osteoblast function has been described for other arboviruses. In case of ZIKV infection, the role of osteoblasts in ZIKV pathogenesis and bone related pathology remains unknown. Here, we study the effect of ZIKV infection on osteoblast differentiation, maturation and function by quantifying activity and gene expression of key biomarkers, using human bone marrow-derived mesenchymal stromal cells (MSCs, osteoblast precursors). MSCs were induced to differentiate into osteoblasts and we found that osteoblasts were highly susceptible to ZIKV infection. While infection did not cause a cytopathic effect, a significant reduction of key osteogenic markers such as ALP, RUNX2, calcium contents and increased expression of IL6 in ZIKV-infected MSCs implicated a delay in osteoblast development and maturation, as compared to uninfected controls. In conclusion, we have developed and characterized a new in vitro model to study the role of bone development in ZIKV pathogenesis, which will help to identify possible new targets for developing therapeutic and preventive measures.
Collapse
Affiliation(s)
- Noreen Mumtaz
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Petra B van den Doel
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Marion P G Koopmans
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Bram C J van der Eerden
- Department of Internal Medicine, Erasmus University Medical Centre, Rotterdam, The Netherlands.
| | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Centre, Rotterdam, The Netherlands.
| |
Collapse
|
81
|
ICR suckling mouse model of Zika virus infection for disease modeling and drug validation. PLoS Negl Trop Dis 2018; 12:e0006848. [PMID: 30356305 PMCID: PMC6218097 DOI: 10.1371/journal.pntd.0006848] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 11/05/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023] Open
Abstract
Background Zika virus (ZIKV) infection causes diseases ranging from acute self-limiting febrile illness to life-threatening Guillain–Barré Syndrome and other neurological disorders in adults. Cumulative evidence suggests an association between ZIKV infection and microcephaly in newborn infants. Given the host-range restrictions of the virus, a susceptible animal model infected by ZIKV must be developed for evaluation of vaccines and antivirals. In this study, we propose a convenient mouse model for analysis of neurological disorders caused by ZIKV. Methodology Six-day-old immunocompetent ICR suckling mice were used in the experiment. Different inoculum virus concentrations, challenge routes, and challenge times were assessed. Viremic dissemination was determined in the liver, spleen, kidney, and brain through Western blot assay, plaque assay, absolute quantification real-time PCR, and histological observation. Azithromycin, a well-characterized anti-ZIKV compound, was used to evaluate the ICR suckling mouse model for antiviral testing. Conclusions Signs of illness and neurological disease and high mortality rate were observed in mice injected with ZIKV intracerebrally (102 to 105) and intraperitoneally (103 to 105). Viremic dissemination was observed in the liver, spleen, kidney, and brain. ZIKV transmitted, rapid replicated, and induced monocyte infiltration into the brain approximately 5 to 6 days post inoculum. Azithromycin conferred protection against ZIKV-caused neurological and life-threatening diseases. The developed model of ZIKV infection and disease can be used for screening drugs against ZIKV and discovering the underlying mechanism of ZIKV pathogenesis. Mosquito-borne Zika virus (ZIKV) is an emerging threat to human health worldwide. In 2007, a ZIKV outbreak was reported in the Yap Island of Micronesia and was the first outbreak outside Africa and Asia. In 2013 and 2014, another ZIKV outbreak was reported in French Polynesia, and more than 28,800 people were infected by ZIKV. In 2015, the first ZIKV outbreak in America was reported in Brazil; the Brazilian Ministry of Health reported a 20-fold increase in cases of neonatal microcephaly, which was geographically and temporally correlated with the ZIKV outbreak. Recent evidence demonstrated that ZIKV infection leads to severe syndromes, such as Guillain–Barré syndrome and microcephaly in adults and infants, respectively. Thus far, anti-ZIKV drugs and vaccines have not been developed yet. Moreover, the underlying mechanism of ZIKV pathogenesis remains unclear. In this study, we propose a small animal model of wild-type ZIKV infection and associated neurological disorders. In the animal model, ZIKV causes signs of illness and neurological disease, potentially emulating the hallmark of ZIKV infection in human. These features can be used to study the underlying mechanism of ZIKV pathogenesis. The newly developed Zika disease model provides an immunocompetent and time saving framework for development of drugs against ZIKV and ZIKV-caused diseases.
Collapse
|
82
|
The celecoxib derivative kinase inhibitor AR-12 (OSU-03012) inhibits Zika virus via down-regulation of the PI3K/Akt pathway and protects Zika virus-infected A129 mice: A host-targeting treatment strategy. Antiviral Res 2018; 160:38-47. [PMID: 30326204 PMCID: PMC7113887 DOI: 10.1016/j.antiviral.2018.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 09/22/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022]
Abstract
Zika virus (ZIKV) is a human-pathogenic flavivirus that has recently emerged as a global public health threat. ZIKV infection may be associated with congenital malformations in infected fetuses and severe neurological and systemic complications in infected adults. There are currently limited treatment options for ZIKV infection. AR-12 (OSU-03012) is a celecoxib derivative cellular kinase inhibitor that has broad-spectrum antiviral activities. In this study, we investigated the antiviral activity and mechanism of AR-12 against ZIKV. We evaluated the in vitro anti-ZIKV activity of AR-12, using cell protection and virus yield reduction assays, in multiple clinically relevant cell lines, and the in vivo treatment effects of AR-12 in a lethal mouse model using type I interferon receptor-deficient A129 mice. AR-12 inhibited ZIKV strains belonging to both the African and Asian/American lineages in Huh-7 and/or neuronal cells. AR12's IC50 against ZIKV was consistently <2 μM in these cells. ZIKV-infected A129 mice treated with intraperitoneally or orally administered AR-12 had significantly higher survival rate (50.0%–83.3% vs 0%, P < 0.05), less body weight loss, and lower blood and tissue ZIKV RNA loads than untreated control A129 mice. These anti-ZIKV effects were likely the results of down-regulation of the PI3K/Akt pathway by AR-12. Clinical trials using the clinically available and broad-spectrum AR-12 as an empirical treatment should be considered especially for patients residing in or returning from areas endemic of ZIKV and other arboviral infections who present with an acute undifferentiated febrile illness. AR-12 (OSU-03012) inhibited the replication of Zika virus strains belonging to both the Asian/American and African lineages. AR-12 inhibited Zika virus replication in multiple cell types in vitro. AR-12 treatment improved clinical and virological outcome of Zika virus-infected type I interferon receptor-deficient mice. AR-12 inhibited Zika virus replication via down-regulation of protein kinase B (Akt).
Collapse
|
83
|
Medlock JM, Hansford KM, Vaux AGC, Cull B, Gillingham E, Leach S. Assessment of the Public Health Threats Posed by Vector-Borne Disease in the United Kingdom (UK). INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2018; 15:E2145. [PMID: 30274268 PMCID: PMC6210260 DOI: 10.3390/ijerph15102145] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 12/21/2022]
Abstract
In recent years, the known distribution of vector-borne diseases in Europe has changed, with much new information also available now on the status of vectors in the United Kingdom (UK). For example, in 2016, the UK reported their first detection of the non-native mosquito Aedes albopictus, which is a known vector for dengue and chikungunya virus. In 2010, Culex modestus, a principal mosquito vector for West Nile virus was detected in large numbers in the Thames estuary. For tick-borne diseases, data on the changing distribution of the Lyme borreliosis tick vector, Ixodes ricinus, has recently been published, at a time when there has been an increase in the numbers of reported human cases of Lyme disease. This paper brings together the latest surveillance data and pertinent research on vector-borne disease in the UK, and its relevance to public health. It highlights the need for continued vector surveillance systems to monitor our native mosquito and tick fauna, as well as the need to expand surveillance for invasive species. It illustrates the importance of maintaining surveillance capacity that is sufficient to ensure accurate and timely disease risk assessment to help mitigate the UK's changing emerging infectious disease risks, especially in a time of climatic and environmental change and increasing global connectivity.
Collapse
Affiliation(s)
- Jolyon M Medlock
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
- Health Protection Research Unit in Environmental Change and Health, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
- Health Protection Research Unit in Emerging and Zoonotic Infections, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | - Kayleigh M Hansford
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
- Health Protection Research Unit in Environmental Change and Health, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | - Alexander G C Vaux
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | - Ben Cull
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | - Emma Gillingham
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
- Health Protection Research Unit in Environmental Change and Health, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| | - Steve Leach
- Medical Entomology Group, Public Health England, Emergency Response Department, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
- Health Protection Research Unit in Emerging and Zoonotic Infections, Porton Down, Salisbury, Wiltshire SP4 0JG, UK.
| |
Collapse
|
84
|
Structurally- and dynamically-driven allostery of the chymotrypsin-like proteases of SARS, Dengue and Zika viruses. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 143:52-66. [PMID: 30217495 PMCID: PMC7111307 DOI: 10.1016/j.pbiomolbio.2018.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/09/2018] [Accepted: 08/24/2018] [Indexed: 01/19/2023]
Abstract
Coronavirus 3C-like and Flavivirus NS2B-NS3 proteases utilize the chymotrypsin fold to harbor their catalytic machineries but also contain additional domains/co-factors. Over the past decade, we aimed to decipher how the extra domains/co-factors mediate the catalytic machineries of SARS 3C-like, Dengue and Zika NS2B-NS3 proteases by characterizing their folding, structures, dynamics and inhibition with NMR, X-ray crystallography and MD simulations, and the results revealed: 1) the chymotrypsin fold of the SARS 3C-like protease can independently fold, while, by contrast, those of Dengue and Zika proteases lack the intrinsic capacity to fold without co-factors. 2) Mutations on the extra domain of SARS 3C-like protease can transform the active catalytic machinery into the inactive collapsed state by structurally-driven allostery. 3) Amazingly, even without detectable structural changes, mutations on the extra domain are sufficient to either inactivate or enhance the catalytic machinery of SARS 3C-like protease by dynamically-driven allostery. 4) Global networks of correlated motions have been identified: for SARS 3C-like protease, N214A inactivates the catalytic machinery by decoupling the network, while STI/A and STIF/A enhance by altering the patterns of the network. The global networks of Dengue and Zika proteases are coordinated by their NS2B-cofactors. 5) Natural products were identified to allosterically inhibit Zika and Dengue proteases through binding a pocket on the back of the active site. Therefore, by introducing extra domains/cofactors, nature develops diverse strategies to regulate the catalytic machinery embedded on the chymotrypsin fold through folding, structurally- and dynamically-driven allostery, all of which might be exploited to develop antiviral drugs.
Collapse
|
85
|
A cross-sectional analysis of Zika virus infection in symptomatic and asymptomatic non-pregnant travellers: Experience of a European reference center during the outbreak in the Americas. Travel Med Infect Dis 2018; 27:107-114. [PMID: 30205195 DOI: 10.1016/j.tmaid.2018.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 08/14/2018] [Accepted: 08/24/2018] [Indexed: 11/20/2022]
Abstract
BACKGROUND Zika virus (ZIKV) infection a concern to travellers because of potential sexual transmission and adverse pregnancy outcomes. OBJECTIVE To describe our experience in diagnosing ZIKV in travellers returning from endemic territories. METHOD Travellers were evaluated for ZIKV at our clinic in a 12-month period during the outbreak, using ZIKV-specific RT-PCR and anti-ZIKV Immunoglobulin M/G ELISA when symptomatic, and ELISA only for asymptomatic travellers, preferably from 20 days after the last exposure. All positive ELISA results were subject to confirmation by Virus Neutralization Testing. We estimated post-test probabilities of ZIKV in asymptomatic travellers. RESULTS Of 462 travellers, 227 reported symptoms and 235 did not. Asymptomatic travellers had similar baseline characteristics, but were younger (median age 31 vs. 33 years, p = 0.01) and had reproductive concerns more often (75.8% vs. 24.2%). ZIKV infection was confirmed in 49 cases: 46/227 (20.3%) were symptomatic and 3/235 (1.3%) asymptomatic. Rash (positive likelihood ratio (LRP) 5.6) and conjunctivitis (LRP 10.8) predicted ZIKV infection. The post-test probability of a negative ELISA-result at 20-25 days was below 0.1%. CONCLUSION ZIKV infection was frequent in symptomatic, but not in asymptomatic travellers. We consider negative ELISA results at 20-25 days after exposure a safe strategy to rule out ZIKV infection. Testing for ZIKV-specific antibodies within this timeframe could be particularly valuable in the management of returning travellers who wish to conceive.
Collapse
|
86
|
Hassert M, Wolf KJ, Schwetye KE, DiPaolo RJ, Brien JD, Pinto AK. CD4+T cells mediate protection against Zika associated severe disease in a mouse model of infection. PLoS Pathog 2018; 14:e1007237. [PMID: 30212537 PMCID: PMC6136803 DOI: 10.1371/journal.ppat.1007237] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV) has gained worldwide attention since it emerged, and a global effort is underway to understand the correlates of protection and develop diagnostics to identify rates of infection. As new therapeutics and vaccine approaches are evaluated in clinical trials, additional effort is focused on identifying the adaptive immune correlates of protection against ZIKV disease. To aid in this endeavor we have begun to dissect the role of CD4+T cells in the protection against neuroinvasive ZIKV disease. We have identified an important role for CD4+T cells in protection, demonstrating that in the absence of CD4+T cells mice have more severe neurological sequela and significant increases in viral titers in the central nervous system (CNS). The transfer of CD4+T cells from ZIKV immune mice protect type I interferon receptor deficient animals from a lethal challenge; showing that the CD4+T cell response is necessary and sufficient for control of ZIKV disease. Using a peptide library spanning the complete ZIKV polyprotein, we identified both ZIKV-encoded CD4+T cell epitopes that initiate immune responses, and ZIKV specific CD4+T cell receptors that recognize these epitopes. Within the ZIKV antigen-specific TCRβ repertoire, we uncovered a high degree of diversity both in response to a single epitope and among different mice responding to a CD4+T cell epitope. Overall this study identifies a novel role for polyfunctional and polyclonal CD4+T cells in providing protection against ZIKV infection and highlights the need for vaccines to develop robust CD4+T cell responses to prevent ZIKV neuroinvasion and limit replication within the CNS.
Collapse
MESH Headings
- Adoptive Transfer
- Amino Acid Sequence
- Animals
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- CD4-Positive T-Lymphocytes/immunology
- Central Nervous System/immunology
- Central Nervous System/virology
- Disease Models, Animal
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Genes, T-Cell Receptor beta
- Humans
- Immunity, Cellular
- Liver/immunology
- Liver/virology
- Lymphocyte Depletion
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptor, Interferon alpha-beta/deficiency
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/immunology
- Viral Vaccines/immunology
- Virus Replication/immunology
- Zika Virus/genetics
- Zika Virus/immunology
- Zika Virus/pathogenicity
- Zika Virus Infection/genetics
- Zika Virus Infection/immunology
- Zika Virus Infection/prevention & control
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, Missouri, United States of America
| | - Kyle J. Wolf
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, Missouri, United States of America
| | - Katherine E. Schwetye
- Department of Pathology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Richard J. DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, Missouri, United States of America
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, Missouri, United States of America
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, Missouri, United States of America
| |
Collapse
|
87
|
Karkhah A, Nouri HR, Javanian M, Koppolu V, Masrour-Roudsari J, Kazemi S, Ebrahimpour S. Zika virus: epidemiology, clinical aspects, diagnosis, and control of infection. Eur J Clin Microbiol Infect Dis 2018; 37:2035-2043. [PMID: 30167886 DOI: 10.1007/s10096-018-3354-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 08/09/2018] [Indexed: 11/24/2022]
Abstract
Zika virus (ZIKV) is an emerging pathogen of huge public health significance to human beings. Although majority of infections are benign with self-limiting symptoms, the recent outbreak has established an association with the increased incidence of some congenital anomalies such as microcephaly. In other words, due to the large extent of the virus and mosquito vectors, the infection has become a thoughtful health problem for human societies, though now, there are no antiviral therapies or vaccines against this virus. In spite of extensive research carried out by scientists, not so much information has been gathered about this viral infection. In the current review, we prepared an overview of the remarkable progress made in understanding about the epidemiology, immunology, clinical presentation, and diagnosis methods of ZIKV infection.
Collapse
Affiliation(s)
- Ahmad Karkhah
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Student Research Committee, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Hamid Reza Nouri
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mostafa Javanian
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Veerendra Koppolu
- Scientist Biopharmaceutical Development Medimmune, Gaithersburg, MD, 20878, USA
| | - Jila Masrour-Roudsari
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Kazemi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
88
|
Abstract
Zika virus (ZIKV) is an arthropod-borne virus that was first isolated in 1947. Since then, multiple outbreaks of ZIKV have been reported in different countries. It is transmitted mostly by Aedes mosquitoes, and the symptoms of fever, joint pain, red eyes, headache, and maculopapular rash closely resemble chikungunya and dengue. The most severe complications of ZIKV infection include the risk of microcephaly and other congenital brain anomalies in infected pregnant women. It is a condition that can be easily prevented with proper education. In this review, we focussed on different aspects of the recent outbreaks of Zika virus disease, including its epidemiological background, transmission routes, presentation, as well as diagnosis and prevention.
Collapse
Affiliation(s)
- Vivian C Agumadu
- Medicine, International University of the Health Sciences School of Medicine, Basseterre, KNA
| | - Kamleshun Ramphul
- Pediatrics, Shanghai Jiao Tong University School of Medicine/Shanghai Xin Hua Hospital, Shanghai, CHN
| |
Collapse
|
89
|
Jackman JA, Cho NJ. Targeting the Achilles Heel of Zika Virus and Other Emerging Viral Pathogens. ADVANCED THERAPEUTICS 2018. [DOI: 10.1002/adtp.201800045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Joshua A. Jackman
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
| | - Nam-Joon Cho
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
- School of Chemical and Biomedical Engineering; Nanyang Technological University; 62 Nanyang Drive Singapore 637459
| |
Collapse
|
90
|
Beltrán-Silva S, Chacón-Hernández S, Moreno-Palacios E, Pereyra-Molina J. Clinical and differential diagnosis: Dengue, chikungunya and Zika. REVISTA MÉDICA DEL HOSPITAL GENERAL DE MÉXICO 2018. [DOI: 10.1016/j.hgmx.2016.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
91
|
da Silva S, Oliveira Silva Martins D, Jardim ACG. A Review of the Ongoing Research on Zika Virus Treatment. Viruses 2018; 10:E255. [PMID: 29758005 PMCID: PMC5977248 DOI: 10.3390/v10050255] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 12/17/2022] Open
Abstract
The Zika fever is an arboviral disease resulting from the infection with Zika virus (ZIKV). The virus is transmitted to humans by the bite of Aedes mosquitos, mainly Aedes aegypti and Aedes albopictus. ZIKV has been detected for decades in African and Asian regions and, since 2007, has spread to other continents; among them, infections are most reported in the Americas. This can be explained by the presence of vectors in highly populated and tropical regions where people are susceptible to contamination. ZIKV has been considered by the World Health Organization a serious public health problem because of the increasing number of cases of congenital malformation and neurological disorders related to its infection, such as microcephaly, Guillain⁻Barré syndrome, meningoencephalitis, and myelitis. There is no vaccine or specific antiviral against ZIKV. The infection is best prevented by avoiding mosquito bite, and the treatment of infected patients is palliative. In this context, the search for efficient antivirals is necessary but remains challenging. Here, we aim to review the molecules that have been described to interfere with ZIKV life cycle and discuss their potential use in ZIKV therapy.
Collapse
Affiliation(s)
- Suely da Silva
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| | - Daniel Oliveira Silva Martins
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, ICBIM, Federal University of Uberlândia, Uberlândia, MG 38405-302, Brazil.
- Genomics Study Laboratory, São Paulo State University, IBILCE, São José do Rio Preto, SP 15054-000, Brazil.
| |
Collapse
|
92
|
Morris G, Barichello T, Stubbs B, Köhler CA, Carvalho AF, Maes M. Zika Virus as an Emerging Neuropathogen: Mechanisms of Neurovirulence and Neuro-Immune Interactions. Mol Neurobiol 2018; 55:4160-4184. [PMID: 28601976 DOI: 10.1007/s12035-017-0635-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023]
Abstract
Zika virus (ZIKV) is an emerging arbovirus of the genus Flaviviridae, which causes a febrile illness and has spread from across the Pacific to the Americas in a short timeframe. Convincing evidence has implicated the ZIKV to incident cases of neonatal microcephaly and a set of neurodevelopmental abnormalities referred to as the congenital Zika virus syndrome. In addition, emerging data points to an association with the ZIKV and the development of the so-called Guillain-Barre syndrome, an acute autoimmune polyneuropathy. Accumulating knowledge suggests that neurovirulent strains of the ZIKV have evolved from less pathogenic lineages of the virus. Nevertheless, mechanisms of neurovirulence and host-pathogen neuro-immune interactions remain incompletely elucidated. This review provides a critical discussion of genetic and structural alterations in the ZIKV which could have contributed to the emergence of neurovirulent strains. In addition, a mechanistic framework of neuro-immune mechanisms related to the emergence of neuropathology after ZIKV infection is discussed. Recent advances in knowledge point to avenues for the development of a putative vaccine as well as novel therapeutic strategies. Nevertheless, there are unique unmet challenges that need to be addressed in this regard. Finally, a research agenda is proposed.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
| | - Tatiana Barichello
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, Denmark Hill, London, SE5 8AZ, UK
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Bishop Hall Lane, Chelmsford, CM1 1SQ, UK
| | - Cristiano A Köhler
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, VIC, 3220, Australia.
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Parana, Brazil.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Revitalis, Waalre, The Netherlands.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
93
|
McLean E, Bhattarai R, Hughes BW, Mahalingam K, Bagasra O. Computational identification of mutually homologous Zika virus miRNAs that target microcephaly genes. Libyan J Med 2018; 12:1304505. [PMID: 28385119 PMCID: PMC5418939 DOI: 10.1080/19932820.2017.1304505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background Zika virus (ZIKV) has been associated with a variety of neuropathologies, including microcephaly. We hypothesize that ZIKV genes activate host microRNAs (miRNAs) causing dysfunctional development of human fetal brains. Objectives/methods A bioinformatics search for miRNA genome-wide binding sites in the prototypic ZIKV (strain MR766) was undertaken to hunt for miRNAs with significant similarities with MCPH genetic sequences responsible for inducing MCHP in human fetal brains. Results Six ZIKV miRNAs were found to share mutual homology with 12 MCPH genetic sequences responsible for inducing MCPH. Noteworthy was miR-1304, which expressed 100% identity to six different MCPH genes. Conclusions We suggest that following infection of fetal neurons ZIKV may modulate the action of various miRNAs, and miR-1304 in particular, resulting in microcephaly.
Collapse
Affiliation(s)
- Ewen McLean
- a Department of Biology, South Carolina Center for Biotechnology , Claflin University , SC , USA
| | - Roshan Bhattarai
- a Department of Biology, South Carolina Center for Biotechnology , Claflin University , SC , USA
| | - Brandon W Hughes
- a Department of Biology, South Carolina Center for Biotechnology , Claflin University , SC , USA
| | - Kuhanandha Mahalingam
- b Information Technology and Department of Computer Science & Mathematics , Claflin University , Orangeburg , SC , USA
| | - Omar Bagasra
- a Department of Biology, South Carolina Center for Biotechnology , Claflin University , SC , USA
| |
Collapse
|
94
|
Stassen L, Armitage CW, van der Heide DJ, Beagley KW, Frentiu FD. Zika Virus in the Male Reproductive Tract. Viruses 2018; 10:v10040198. [PMID: 29659541 PMCID: PMC5923492 DOI: 10.3390/v10040198] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 01/07/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) are resurging across the globe. Zika virus (ZIKV) has caused significant concern in recent years because it can lead to congenital malformations in babies and Guillain-Barré syndrome in adults. Unlike other arboviruses, ZIKV can be sexually transmitted and may persist in the male reproductive tract. There is limited information regarding the impact of ZIKV on male reproductive health and fertility. Understanding the mechanisms that underlie persistent ZIKV infections in men is critical to developing effective vaccines and therapies. Mouse and macaque models have begun to unravel the pathogenesis of ZIKV infection in the male reproductive tract, with the testes and prostate gland implicated as potential reservoirs for persistent ZIKV infection. Here, we summarize current knowledge regarding the pathogenesis of ZIKV in the male reproductive tract, the development of animal models to study ZIKV infection at this site, and prospects for vaccines and therapeutics against persistent ZIKV infection.
Collapse
Affiliation(s)
- Liesel Stassen
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Charles W Armitage
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - David J van der Heide
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Kenneth W Beagley
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| | - Francesca D Frentiu
- Institute of Health and Biomedical Innovation, and School of Biomedical Sciences, Queensland University of Technology, Brisbane 4006, Queensland, Australia.
| |
Collapse
|
95
|
Abstract
Zika virus (ZIKV) infection is an emergent worldwide public health problem. Historically, 84 countries have reported vector-borne ZIKV transmission, 61 of which report on-going transmission. It is a Flavivirus transmitted through arthropods belonging to the Aedes genus. Since 2015, ZIKV infections have increased dramatically; with 1.3 million people infected during 2015 in Brazil alone. This paper's objective is to highlight the conjectural epidemiological points of the virus' dissemination. The digital archives Pubmed, MEDLINE, EMBASE and Cochrane were searched for papers that assessed aspects of ZIKV transmission and epidemiology. The first isolation occurred in Uganda in 1947. Since then, important outbreaks were documented globally. Consequently, an emergent public health problem arose from a rapidly increasing incidence and its association with the development of neurological diseases such as microcephaly and Guillain-Barré syndrome. Key factors in the successful containment of outbreaks include surveillance of mosquitos in the neighbourhood, an early mosquito control treatment, an assertive information campaign, and the involvement of the local population and healthcare workers. As such, while ZIKV seems to be spreading globally in a similar manner to other arboviruses, such as Dengue and Chikungunya viruses, it can also be rapidly contained due to the pre-existing availability of necessary resources and regulatory tools as control measures. This review aims to provide a description of those characteristics of ZIKV infection that may be useful in the construction of effective outbreak control strategies.
Collapse
|
96
|
Kim YH, Lee J, Kim YE, Chong CK, Pinchemel Y, Reisdörfer F, Coelho JB, Dias RF, Bae PK, Gusmão ZPM, Ahn HJ, Nam HW. Development of a Rapid Diagnostic Test Kit to Detect IgG/IgM Antibody against Zika Virus Using Monoclonal Antibodies to the Envelope and Non-structural Protein 1 of the Virus. THE KOREAN JOURNAL OF PARASITOLOGY 2018. [PMID: 29529852 PMCID: PMC5858665 DOI: 10.3347/kjp.2018.56.1.61] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We developed a Rapid Diagnostic Test (RDT) kit for detecting IgG/IgM antibodies against Zika virus (ZIKV) using monoclonal antibodies to the envelope (E) and non-structural protein 1 (NS1) of ZIKV. These proteins were produced using baculovirus expression vector with Sf9 cells. Monoclonal antibodies J2G7 to NS1 and J5E1 to E protein were selected and conjugated with colloidal gold to produce the Zika IgG/IgM RDT kit (Zika RDT). Comparisons with ELISA, plaque reduction neutralization test (PRNT), and PCR were done to investigate the analytical sensitivity of Zika RDT, which resulted in 100% identical results. Sensitivity and specificity of Zika RDT in a field test was determined using positive and negative samples from Brazil and Korea. The diagnostic accuracy of Zika RDT was fairly high; sensitivity and specificity for IgG was 99.0 and 99.3%, respectively, while for IgM it was 96.7 and 98.7%, respectively. Cross reaction with dengue virus was evaluated using anti-Dengue Mixed Titer Performance Panel (PVD201), in which the Zika RDT showed cross-reactions with DENV in 16.7% and 5.6% in IgG and IgM, respectively. Cross reactions were not observed with West Nile, yellow fever, and hepatitis C virus infected sera. Zika RDT kit is very simple to use, rapid to assay, and very sensitive, and highly specific. Therefore, it would serve as a choice of method for point-of-care diagnosis and large scale surveys of ZIKV infection under clinical or field conditions worldwide in endemic areas.
Collapse
Affiliation(s)
- Yeong Hoon Kim
- Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | | | | | | | | | | | | | | | - Pan Kee Bae
- BioNano Health Guard Research Center, Daejeon 34141, Korea
| | | | - Hye-Jin Ahn
- Department of Parasitology, College of Medicine, Catholic University of Korea, Seoul 06591, Korea
| | - Ho-Woo Nam
- Department of Parasitology, College of Medicine, Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
97
|
Alvarado-Socarras JL, Idrovo ÁJ, Contreras-García GA, Rodriguez-Morales AJ, Audcent TA, Mogollon-Mendoza AC, Paniz-Mondolfi A. Congenital microcephaly: A diagnostic challenge during Zika epidemics. Travel Med Infect Dis 2018; 23:14-20. [PMID: 29471046 DOI: 10.1016/j.tmaid.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/02/2018] [Accepted: 02/13/2018] [Indexed: 01/01/2023]
Abstract
The multiple, wide and diverse etiologies of congenital microcephaly are complex and multifactorial. Recent advances in genetic testing have improved understanding of novel genetic causes of congenital microcephaly. The recent Zika virus (ZIKV) epidemic in Latin America has highlighted the need for a better understanding of the underlying pathological mechanisms of microcephaly including both infectious and non-infectious causes. The diagnostic approach to microcephaly needs to include potential infectious and genetic etiologies, as well as environmental in-utero exposures such as alcohol, toxins, and medications. Emerging genetic alterations linked to microcephaly include abnormal mitotic microtubule spindle structure and abnormal function of centrosomes. We discuss the diagnostic challenge of congenital microcephaly in the context of understanding the links with ZIKV emergence as a new etiological factor involved in this birth defect.
Collapse
Affiliation(s)
- Jorge L Alvarado-Socarras
- Neonatal Unit, Department of Pediatrics, Fundación Cardiovascular de Colombia, Floridablanca, Santander, Colombia; Organización Latinoamericana para el Fomento de la Investigación en Salud (OLFIS), Bucaramanga, Santander, Colombia; Colombian Collaborative Network on Zika (RECOLZIKA), Pereira, Risaralda, Colombia
| | - Álvaro J Idrovo
- Public Health Department, School of Medicine, Universidad Industrial de Santander, Bucaramanga, Colombia
| | | | - Alfonso J Rodriguez-Morales
- Organización Latinoamericana para el Fomento de la Investigación en Salud (OLFIS), Bucaramanga, Santander, Colombia; Colombian Collaborative Network on Zika (RECOLZIKA), Pereira, Risaralda, Colombia; Public Health and Infection Research Group, Faculty of Health Sciences, Universidad Tecnológica de Pereira, Pereira, Risaralda, Colombia.
| | - Tobey A Audcent
- Children's Hospital of Eastern Ontario, 401 Smyth Rd, Ottawa, ON, K1H 8L1, Canada
| | - Adriana C Mogollon-Mendoza
- Infectious Diseases Research Incubator and the Zoonosis and Emerging Pathogens Regional Collaborative Network, Venezuela; Health Sciences Department, College of Medicine, Universidad Centroccidental Lisandro Alvarado, Barquisimeto, Lara, Venezuela
| | - Alberto Paniz-Mondolfi
- IDB Biomedical Research Center, Department of Infectious Diseases and Tropical Medicine/Infectious Diseases Pathology Laboratory (IDB), Barquisimeto, Venezuela; Directorate of Health, Instituto Venezolano de Los Seguros Sociales (IVSS), Caracas, Dtto. Capital, Venezuela
| |
Collapse
|
98
|
Ramos CL, Moreno-Carvalho OA, Nascimento-Carvalho CM. Cerebrospinal fluid aspects of neonates with or without microcephaly born to mothers with gestational Zika virus clinical symptoms. J Infect 2018; 76:563-569. [PMID: 29432825 DOI: 10.1016/j.jinf.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 01/26/2018] [Accepted: 02/03/2018] [Indexed: 11/24/2022]
Affiliation(s)
- Clara L Ramos
- Bahiana School of Medicine, Bahiana Foundation for Science Development, Salvador, 40295-050, Brazil.
| | | | - Cristiana M Nascimento-Carvalho
- Post-graduation Program in Health Sciences and Department of Paediatrics, Federal University of Bahia School of Medicine, Salvador, 40025-010, Brazil; Post-graduation Program in Health Sciences and Department of Paediatrics, Federal University of Bahia School of Medicine, Salvador, 40025-010, BrazilBrazilian Council for Scientific and Technological Development, Brazilia, Brazil
| |
Collapse
|
99
|
Singh RK, Dhama K, Khandia R, Munjal A, Karthik K, Tiwari R, Chakraborty S, Malik YS, Bueno-Marí R. Prevention and Control Strategies to Counter Zika Virus, a Special Focus on Intervention Approaches against Vector Mosquitoes-Current Updates. Front Microbiol 2018; 9:87. [PMID: 29472902 PMCID: PMC5809424 DOI: 10.3389/fmicb.2018.00087] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/15/2018] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) is the most recent intruder that acquired the status of global threat creating panic and frightening situation to public owing to its rapid spread, attaining higher virulence and causing complex clinical manifestations including microcephaly in newborns and Guillain Barré Syndrome. Alike other flaviviruses, the principal mode of ZIKV transmission is by mosquitoes. Advances in research have provided reliable diagnostics for detecting ZIKV infection, while several drug/therapeutic targets and vaccine candidates have been identified recently. Despite these progresses, currently there is neither any effective drug nor any vaccine available against ZIKV. Under such circumstances and to tackle the problem at large, control measures of which mosquito population control need to be strengthened following appropriate mechanical, chemical, biological and genetic control measures. Apart from this, several other known modes of ZIKV transmission which have gained importance in recent past such as intrauterine, sexual intercourse, and blood-borne spread need to be checked and kept under control by adopting appropriate precautions and utmost care during sexual intercourse, blood transfusion and organ transplantation. The virus inactivation by pasteurization, detergents, chemicals, and filtration can effectively reduce viral load in plasma-derived medicinal products. Added to this, strengthening of the surveillance and monitoring of ZIKV as well as avoiding travel to Zika infected areas would aid in keeping viral infection under check. Here, we discuss the salient advances in the prevention and control strategies to combat ZIKV with a focus on highlighting various intervention approaches against the vector mosquitoes of this viral pathogen along with presenting an overview regarding human intervention measures to counter other modes of ZIKV transmission and spread. Additionally, owing to the success of vaccines for a number of infections globally, a separate section dealing with advances in ZIKV vaccines and transmission blocking vaccines has also been included.
Collapse
Affiliation(s)
- Raj K Singh
- ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, UP Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan, Mathura, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, Agartala, India
| | - Yashpal S Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, India
| | - Rubén Bueno-Marí
- Laboratorios Lokímica, Departamento de Investigación y Desarrollo (I+D), Valencia, Spain
| |
Collapse
|
100
|
Mohr EL, Block LN, Newman CM, Stewart LM, Koenig M, Semler M, Breitbach ME, Teixeira LBC, Zeng X, Weiler AM, Barry GL, Thoong TH, Wiepz GJ, Dudley DM, Simmons HA, Mejia A, Morgan TK, Salamat MS, Kohn S, Antony KM, Aliota MT, Mohns MS, Hayes JM, Schultz-Darken N, Schotzko ML, Peterson E, Capuano S, Osorio JE, O’Connor SL, Friedrich TC, O’Connor DH, Golos TG. Ocular and uteroplacental pathology in a macaque pregnancy with congenital Zika virus infection. PLoS One 2018; 13:e0190617. [PMID: 29381706 PMCID: PMC5790226 DOI: 10.1371/journal.pone.0190617] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/18/2017] [Indexed: 12/03/2022] Open
Abstract
Congenital Zika virus (ZIKV) infection impacts fetal development and pregnancy outcomes. We infected a pregnant rhesus macaque with a Puerto Rican ZIKV isolate in the first trimester. The pregnancy was complicated by preterm premature rupture of membranes (PPROM), intraamniotic bacterial infection and fetal demise 49 days post infection (gestational day 95). Significant pathology at the maternal-fetal interface included acute chorioamnionitis, placental infarcts, and leukocytoclastic vasculitis of the myometrial radial arteries. ZIKV RNA was disseminated throughout fetal tissues and maternal immune system tissues at necropsy, as assessed by quantitative RT-PCR for viral RNA. Replicating ZIKV was identified in fetal tissues, maternal uterus, and maternal spleen by fluorescent in situ hybridization for viral replication intermediates. Fetal ocular pathology included a choroidal coloboma, suspected anterior segment dysgenesis, and a dysplastic retina. This is the first report of ocular pathology and prolonged viral replication in both maternal and fetal tissues following congenital ZIKV infection in a rhesus macaque. PPROM followed by fetal demise and severe pathology of the visual system have not been described in macaque congenital ZIKV infection previously. While this case of ZIKV infection during pregnancy was complicated by bacterial infection with PPROM, the role of ZIKV on this outcome cannot be precisely defined, and further nonhuman primate studies will determine if increased risk for PPROM or other adverse pregnancy outcomes are associated with congenital ZIKV infection.
Collapse
Affiliation(s)
- Emma L. Mohr
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail: (ELM); (TGG)
| | - Lindsey N. Block
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Christina M. Newman
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Laurel M. Stewart
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michelle Koenig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Matthew Semler
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Meghan E. Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Leandro B. C. Teixeira
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland, United States of America
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gabrielle L. Barry
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Troy H. Thoong
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Gregory J. Wiepz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dawn M. Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Heather A. Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Andres Mejia
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Terry K. Morgan
- Departments of Pathology and Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - M. Shahriar Salamat
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Sarah Kohn
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kathleen M. Antony
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Matthew T. Aliota
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Mariel S. Mohns
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jennifer M. Hayes
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Nancy Schultz-Darken
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Michele L. Schotzko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Eric Peterson
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Saverio Capuano
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jorge E. Osorio
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Thomas C. Friedrich
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Thaddeus G. Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail: (ELM); (TGG)
| |
Collapse
|