901
|
Maestre AM, Fernandez-Sesma A. ZIKV Strains' Different Phenotypes in Human Neural Cells Could be a Hint for the Emergence of the New Clinical Neurological Outcomes. EBioMedicine 2016; 13:35-36. [PMID: 27765639 PMCID: PMC5264277 DOI: 10.1016/j.ebiom.2016.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Ana M Maestre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Ana Fernandez-Sesma
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, United States; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, United States.
| |
Collapse
|
902
|
Muffat J, Li Y, Jaenisch R. CNS disease models with human pluripotent stem cells in the CRISPR age. Curr Opin Cell Biol 2016; 43:96-103. [PMID: 27768957 DOI: 10.1016/j.ceb.2016.10.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/19/2016] [Accepted: 10/02/2016] [Indexed: 12/31/2022]
Abstract
In vitro differentiation of human pluripotent stem cells provides a systematic platform to investigate the physiological development and function of the human nervous system, as well as the etiology and consequence when these processes go awry. Recent development in three-dimensional (3D) organotypic culture systems allows modeling of the complex structure formation of the human CNS, and the intricate interactions between various resident neuronal and glial cell types. Combined with an ever-expanding genome editing and regulation toolkit such as CRISPR/Cas9, it is now a possibility to study human neurological disease in the relevant molecular, cellular and anatomical context. In this article, we review recent progress in 3D neural culture and the implications for disease modeling.
Collapse
Affiliation(s)
- Julien Muffat
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States
| | - Yun Li
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, 9Cambridge Center, Cambridge, MA 02142, United States.
| |
Collapse
|
903
|
El Costa H, Gouilly J, Mansuy JM, Chen Q, Levy C, Cartron G, Veas F, Al-Daccak R, Izopet J, Jabrane-Ferrat N. ZIKA virus reveals broad tissue and cell tropism during the first trimester of pregnancy. Sci Rep 2016; 6:35296. [PMID: 27759009 PMCID: PMC5069472 DOI: 10.1038/srep35296] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022] Open
Abstract
The outbreak of the Zika Virus (ZIKV) and its association with fetal abnormalities have raised worldwide concern. However, the cellular tropism and the mechanisms of ZIKV transmission to the fetus during early pregnancy are still largely unknown. Therefore, we ex vivo modeled the ZIKV transmission at the maternal-fetal interface using organ culture from first trimester pregnancy samples. Here, we provide evidence that ZIKV strain circulating in Brazil infects and damages tissue architecture of the maternal decidua basalis, the fetal placenta and umbilical cord. We also show that ZIKV replicates differentially in a wide range of maternal and fetal cells, including decidual fibroblasts and macrophages, trophoblasts, Hofbauer cells as well as umbilical cord mesenchymal stem cells. The striking cellular tropism of ZIKV and its cytopathic-induced tissue injury during the first trimester of pregnancy could provide an explanation for the irreversible congenital damages.
Collapse
Affiliation(s)
- Hicham El Costa
- CPTP, INSERM U1043, CNRS UMR5282, Université Toulouse III, 31024 Toulouse, France.,Laboratoire de Virologie, IFB, CHU Toulouse, 31059 Toulouse, France
| | - Jordi Gouilly
- CPTP, INSERM U1043, CNRS UMR5282, Université Toulouse III, 31024 Toulouse, France
| | | | - Qian Chen
- CPTP, INSERM U1043, CNRS UMR5282, Université Toulouse III, 31024 Toulouse, France
| | - Claude Levy
- Service de Gynécologie-Obstétrique, Clinique Sarrus-Teinturiers, 31300 Toulouse, France
| | - Géraldine Cartron
- Service de Gynécologie-Obstétrique, CHU Toulouse, 31059 Toulouse, France
| | - Francisco Veas
- IRD, UMR-Ministère de la Défense, Faculté de Pharmacie, Université de Montpellier, 34094 Montpellier, France
| | - Reem Al-Daccak
- INSERM UMRS976, Université Paris Diderot, Hôpital Saint-Louis, 75010 Paris, France
| | - Jacques Izopet
- CPTP, INSERM U1043, CNRS UMR5282, Université Toulouse III, 31024 Toulouse, France.,Laboratoire de Virologie, IFB, CHU Toulouse, 31059 Toulouse, France
| | | |
Collapse
|
904
|
Gruba N, Rodriguez Martinez JI, Grzywa R, Wysocka M, Skoreński M, Burmistrz M, Łęcka M, Lesner A, Sieńczyk M, Pyrć K. Substrate profiling of Zika virus NS2B-NS3 protease. FEBS Lett 2016; 590:3459-3468. [PMID: 27714789 DOI: 10.1002/1873-3468.12443] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 01/05/2023]
Abstract
Zika virus (ZIKV), isolated from macaques in Uganda in 1947, was not considered to be a dangerous human pathogen. However, this view has recently changed as ZIKV infections are now associated with serious pathological disorders including microcephaly and Guillain-Barré syndrome. Similar to other viruses in the Flaviviridae family, ZIKV expresses the serine protease NS3 which is responsible for viral protein processing and replication. Herein, we report the expression of an active NS3pro domain fused with the NS2B cofactor (NS2BLN NS3pro ) in a prokaryotic expression system and profile its specificity for synthesized FRET-type substrate libraries. Our findings pave way for screening potential intracellular substrates of NS3 and for developing specific inhibitors of this ZIKV protease.
Collapse
Affiliation(s)
- Natalia Gruba
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, Poland
| | | | - Renata Grzywa
- Division of Medicinal Chemistry and Microbiology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Magdalena Wysocka
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, Poland
| | - Marcin Skoreński
- Division of Medicinal Chemistry and Microbiology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Michał Burmistrz
- Department of Microbiology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Maria Łęcka
- Division of Medicinal Chemistry and Microbiology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Adam Lesner
- Department of Biochemistry, Faculty of Chemistry, University of Gdansk, Poland.
| | - Marcin Sieńczyk
- Division of Medicinal Chemistry and Microbiology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland.
| | - Krzysztof Pyrć
- Department of Microbiology, Faculty of Biochemistry Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland. .,Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
905
|
Goodfellow FT, Tesla B, Simchick G, Zhao Q, Hodge T, Brindley MA, Stice SL. Zika Virus Induced Mortality and Microcephaly in Chicken Embryos. Stem Cells Dev 2016; 25:1691-1697. [PMID: 27627457 DOI: 10.1089/scd.2016.0231] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The explosive spread of the Zika virus (ZIKV) through South and Central America has been linked to an increase in congenital birth defects, specifically microcephaly. Representative rodent models for investigating infections include direct central nervous system (CNS) injections late in pregnancy and transplacental transmission in immunodeficient mice. Microcephaly in humans may be the result of infection occurring early in pregnancy, therefore recapitulating that the human course of ZIKV infection should include normal embryo exposed to ZIKV during the first trimester. In ovo development of the chicken embryo closely mirrors human fetal neurodevelopment and, as a comparative model, could provide key insights into both temporal and pathophysiological effects of ZIKV. Chick embryos were directly infected early and throughout incubation with ZIKV isolated from a Mexican mosquito in January 2016. High doses of virus caused embryonic lethality. In a subset of lower dosed embryos, replicating ZIKV was present in various organs, including the CNS, throughout development. Surviving ZIKV-infected embryos presented a microcephaly-like phenotype. Chick embryos were longitudinally monitored by magnetic resonance imaging that documented CNS structural malformations, including enlarged ventricles (30% increase) and stunted cortical growth (decreased telencephalon by 18%, brain stem by 32%, and total brain volume by 18%), on both embryonic day 15 (E15) and E20 of development. ZIKV-induced microcephaly was observed with inoculations of as few as 2-20 viral particles. The chick embryo model presented ZIKV embryonic lethal effects and progressive CNS damage similar to microcephaly.
Collapse
Affiliation(s)
- Forrest T Goodfellow
- 1 Department of Animal and Dairy Science, Interdisciplinary Toxicology Program, Regenerative Bioscience Center, College of Agriculture and Environmental Science, University of Georgia , Athens, Georgia
| | - Blanka Tesla
- 2 Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia , Athens, Georgia
| | - Gregory Simchick
- 3 Bioimaging Research Center and Department of Physics and Astronomy, University of Georgia , Athens, Georgia
| | - Qun Zhao
- 3 Bioimaging Research Center and Department of Physics and Astronomy, University of Georgia , Athens, Georgia
| | - Thomas Hodge
- 2 Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia , Athens, Georgia
| | - Melinda A Brindley
- 4 Department of Infectious Diseases, Population Health, Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia , Athens, Georgia
| | - Steven L Stice
- 5 Department of Animal and Dairy Science, Regenerative Bioscience Center, College of Agriculture and Environmental Science, University of Georgia , Athens, Georgia
| |
Collapse
|
906
|
Pascoalino BS, Courtemanche G, Cordeiro MT, Gil LHVG, Freitas-Junior L. Zika antiviral chemotherapy: identification of drugs and promising starting points for drug discovery from an FDA-approved library. F1000Res 2016; 5:2523. [PMID: 27909576 PMCID: PMC5112578 DOI: 10.12688/f1000research.9648.1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/06/2016] [Indexed: 12/14/2022] Open
Abstract
Background The recent epidemics of Zika virus (ZIKV) implicated it as the cause of serious and potentially lethal congenital conditions such microcephaly and other central nervous system defects, as well as the development of the Guillain-Barré syndrome in otherwise healthy patients. Recent findings showed that anti-Dengue antibodies are capable of amplifying ZIKV infection by a mechanism similar to antibody-dependent enhancement, increasing the severity of the disease. This scenario becomes potentially catastrophic when the global burden of Dengue and the advent of the newly approved anti-Dengue vaccines in the near future are taken into account. Thus, antiviral chemotherapy should be pursued as a priority strategy to control the spread of the virus and prevent the complications associated with Zika. Methods Here we describe a fast and reliable cell-based, high-content screening assay for discovery of anti-ZIKV compounds. This methodology has been used to screen the National Institute of Health Clinical Collection compound library, a small collection of FDA-approved drugs. Results and conclusion From 725 FDA-approved compounds triaged, 29 (4%) were found to have anti-Zika virus activity, of which 22 had confirmed (76% of confirmation) by dose-response curves. Five candidates presented selective activity against ZIKV infection and replication in a human cell line. These hits have abroad spectrum of chemotypes and therapeutic uses, offering valuable opportunities for selection of leads for antiviral drug discovery.
Collapse
Affiliation(s)
- Bruno S. Pascoalino
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas-SP, 10000, Brazil
- Present Address: Instituto Butantan, São Paulo-SP, 1500, Brazil
| | | | - Marli T. Cordeiro
- Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz -Fiocruz, Recife/PE, Brazil
| | - Laura H. V. G. Gil
- Centro de Pesquisas Aggeu Magalhães, Fundação Oswaldo Cruz -Fiocruz, Recife/PE, Brazil
| | - Lucio Freitas-Junior
- Laboratório Nacional de Biociências, Centro Nacional de Pesquisa em Energia e Materiais, Campinas-SP, 10000, Brazil
- Present Address: Instituto Butantan, São Paulo-SP, 1500, Brazil
| |
Collapse
|
907
|
Shao Q, Herrlinger S, Yang SL, Lai F, Moore JM, Brindley MA, Chen JF. Zika virus infection disrupts neurovascular development and results in postnatal microcephaly with brain damage. Development 2016; 143:4127-4136. [PMID: 27729407 DOI: 10.1242/dev.143768] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Abstract
Zika virus (ZIKV) infection of pregnant women can result in fetal brain abnormalities. It has been established that ZIKV disrupts neural progenitor cells (NPCs) and leads to embryonic microcephaly. However, the fate of other cell types in the developing brain and their contributions to ZIKV-associated brain abnormalities remain largely unknown. Using intracerebral inoculation of embryonic mouse brains, we found that ZIKV infection leads to postnatal growth restriction including microcephaly. In addition to cell cycle arrest and apoptosis of NPCs, ZIKV infection causes massive neuronal death and axonal rarefaction, which phenocopy fetal brain abnormalities in humans. Importantly, ZIKV infection leads to abnormal vascular density and diameter in the developing brain, resulting in a leaky blood-brain barrier (BBB). Massive neuronal death and BBB leakage indicate brain damage, which is further supported by extensive microglial activation and astrogliosis in virally infected brains. Global gene analyses reveal dysregulation of genes associated with immune responses in virus-infected brains. Thus, our data suggest that ZIKV triggers a strong immune response and disrupts neurovascular development, resulting in postnatal microcephaly with extensive brain damage.
Collapse
Affiliation(s)
- Qiang Shao
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Stephanie Herrlinger
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Si-Lu Yang
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| | - Fan Lai
- University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Department of Human Genetics, Miami, FL 33136, USA
| | - Julie M Moore
- Department of Infectious Diseases and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA 30602, USA
| | - Melinda A Brindley
- Department of Infectious Diseases, Department of Population Health and Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - Jian-Fu Chen
- Department of Genetics, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
908
|
Huang WC, Abraham R, Shim BS, Choe H, Page DT. Zika virus infection during the period of maximal brain growth causes microcephaly and corticospinal neuron apoptosis in wild type mice. Sci Rep 2016; 6:34793. [PMID: 27713505 PMCID: PMC5054421 DOI: 10.1038/srep34793] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 09/16/2016] [Indexed: 12/31/2022] Open
Abstract
Zika virus (ZIKV) infection in pregnant women has been established as a cause of microcephaly in newborns. Here we test the hypothesis that neurodevelopmental stages when the brain is undergoing rapid growth are particularly vulnerable to the effects of ZIKV infection. We injected ZIKV intracranially into wild type C57BL/6 mice at two different time points: early postnatal development, when the brain is growing at its maximal rate, and at weaning, when the brain has largely reached adult size. Both time points showed widespread immunoreactivity for ZIKV and cleaved caspase 3 (CC3, a marker of apoptosis) throughout the brain. However, in early postnatal ZIKV injected mice, some brain areas and cell types display particularly large increases in apoptosis that we did not observe in older animals. Corticospinal pyramidal neurons, a cell type implicated in human microcephaly associated with ZIKV infection, are an example of one such cell type. Proliferating cells in the ventricular zone stem cell compartment are also depleted. These findings are consistent with the hypothesis that periods of rapid brain growth are especially susceptible to neurodevelopmental effects of ZIKV infection, and establish a valuable model to investigate mechanisms underlying neurodevelopmental effects of ZIKV infection and explore candidate therapeutics.
Collapse
Affiliation(s)
- Wen-Chin Huang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,The Doctoral Program in Chemical and Biological Sciences at The Scripps Research Institute, Jupiter, Florida, USA
| | - Rachy Abraham
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Byoung-Shik Shim
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Hyeryun Choe
- Department of Immunology and Microbial Science, The Scripps Research Institute, Jupiter, Florida, USA
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,The Doctoral Program in Chemical and Biological Sciences at The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
909
|
Abstract
CONTEXT -As the number of Zika virus (ZIKV) infections continues to grow, so, too, does the spectrum of recognized clinical disease, in both adult and congenital infections. Defining the tissue pathology associated with the various disease manifestations provides insight into pathogenesis and diagnosis, and potentially future prevention and treatment, of ZIKV infections. OBJECTIVE -To summarize the syndromes and pathology associated with ZIKV infection, the implications of pathologic findings in the pathogenesis of ZIKV disease, and the use of pathology specimens for diagnosis of ZIKV infection. DATA SOURCES -The major sources of information for this review were published articles obtained from PubMed and pathologic findings from cases submitted to the Infectious Diseases Pathology Branch at the Centers for Disease Control and Prevention. CONCLUSIONS -Pathologic findings associated with ZIKV infection are characteristic but not specific. In congenital Zika syndrome, tissue pathology is due to direct viral infection of neural structures, whereas in Guillain-Barré syndrome, pathology is likely due to a postviral, aberrant host-directed immune response. Both fetal and placental pathology specimens are useful for ZIKV diagnosis by molecular and immunohistochemical assays; however, the implications of ZIKV detection in placentas from second- and third-trimester normal live births are unclear, as the potential postnatal effects of late gestational exposure remain to be seen.
Collapse
Affiliation(s)
| | | | - Sherif R Zaki
- From the Infectious Diseases Pathology Branch, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
910
|
Xu M, Lee EM, Wen Z, Cheng Y, Huang WK, Qian X, Tcw J, Kouznetsova J, Ogden SC, Hammack C, Jacob F, Nguyen HN, Itkin M, Hanna C, Shinn P, Allen C, Michael SG, Simeonov A, Huang W, Christian KM, Goate A, Brennand KJ, Huang R, Xia M, Ming GL, Zheng W, Song H, Tang H. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat Med 2016; 22:1101-1107. [PMID: 27571349 PMCID: PMC5386783 DOI: 10.1038/nm.4184] [Citation(s) in RCA: 514] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
In response to the current global health emergency posed by the Zika virus (ZIKV) outbreak and its link to microcephaly and other neurological conditions, we performed a drug repurposing screen of ∼6,000 compounds that included approved drugs, clinical trial drug candidates and pharmacologically active compounds; we identified compounds that either inhibit ZIKV infection or suppress infection-induced caspase-3 activity in different neural cells. A pan-caspase inhibitor, emricasan, inhibited ZIKV-induced increases in caspase-3 activity and protected human cortical neural progenitors in both monolayer and three-dimensional organoid cultures. Ten structurally unrelated inhibitors of cyclin-dependent kinases inhibited ZIKV replication. Niclosamide, a category B anthelmintic drug approved by the US Food and Drug Administration, also inhibited ZIKV replication. Finally, combination treatments using one compound from each category (neuroprotective and antiviral) further increased protection of human neural progenitors and astrocytes from ZIKV-induced cell death. Our results demonstrate the efficacy of this screening strategy and identify lead compounds for anti-ZIKV drug development.
Collapse
Affiliation(s)
- Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Emily M Lee
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Zhexing Wen
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Wei-Kai Huang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xuyu Qian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Julia Tcw
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jennifer Kouznetsova
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarah C Ogden
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Christy Hammack
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Fadi Jacob
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ha Nam Nguyen
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Misha Itkin
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine Hanna
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Paul Shinn
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Chase Allen
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Samuel G Michael
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenwei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Kimberly M Christian
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristen J Brennand
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Guo-Li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Biomedical Engineering Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
911
|
da Silva SR, Gao SJ. Zika virus update II: Recent development of animal models-Proofs of association with human pathogenesis. J Med Virol 2016; 88:1657-8. [PMID: 27208545 PMCID: PMC5240632 DOI: 10.1002/jmv.24582] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2016] [Indexed: 11/10/2022]
Abstract
Three recent studies in pregnant mice and one ongoing study in rhesus macaques evaluating the effect of ZIKV infection have provided important information about maternal-fetus transmission and ZIKV-related pathogenesis, confirming a causal role of ZIKV in neurological problems observed in humans. Here, we present an update of these works published in the past few weeks. J. Med. Virol. 88:1657-1658, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Suzane Ramos da Silva
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Shou-Jiang Gao
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
912
|
Li XF, Dong HL, Huang XY, Qiu YF, Wang HJ, Deng YQ, Zhang NN, Ye Q, Zhao H, Liu ZY, Fan H, An XP, Sun SH, Gao B, Fa YZ, Tong YG, Zhang FC, Gao GF, Cao WC, Shi PY, Qin CF. Characterization of a 2016 Clinical Isolate of Zika Virus in Non-human Primates. EBioMedicine 2016; 12:170-177. [PMID: 27693104 PMCID: PMC5078627 DOI: 10.1016/j.ebiom.2016.09.022] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 09/10/2016] [Accepted: 09/21/2016] [Indexed: 11/16/2022] Open
Abstract
Animal models are critical to understand disease and to develop countermeasures for the ongoing epidemics of Zika virus (ZIKV). Here we report a non-human primate model using a 2016 contemporary clinical isolate of ZIKV. Upon subcutaneous inoculation, rhesus macaques developed fever and viremia, with robust excretion of ZIKV RNA in urine, saliva, and lacrimal fluid. Necropsy of two infected animals revealed that systematic infections involving central nervous system and visceral organs were established at the acute phrase. ZIKV initially targeted the intestinal tracts, spleen, and parotid glands, and retained in spleen and lymph nodes till 10days post infection. ZIKV-specific immune responses were readily induced in all inoculated animals. The non-human primate model described here provides a valuable platform to study ZIKV pathogenesis and to evaluate vaccine and therapeutics.
Collapse
Affiliation(s)
- Xiao-Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hao-Long Dong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xing-Yao Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Ye-Feng Qiu
- Laboratory Animal Center, Academy of Military Medical Science, Beijing 100071, China
| | - Hong-Jiang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yong-Qiang Deng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Na-Na Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Guangxi Medical University, Xining 530021, China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hui Zhao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Zhong-Yu Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hang Fan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Xiao-Ping An
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Shi-Hui Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Bo Gao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yun-Zhi Fa
- Laboratory Animal Center, Academy of Military Medical Science, Beijing 100071, China
| | - Yi-Gang Tong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Fu-Chun Zhang
- Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - George F Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wu-Chun Cao
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, Department of Pharmacology and Toxicology, Sealy Center for Structural Biology & Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Guangxi Medical University, Xining 530021, China.
| |
Collapse
|
913
|
Pyke AT, Warrilow D. Archival Collections are Important in the Study of the Biology, Diversity, and Evolution of Arboviruses. Evol Bioinform Online 2016; 12:27-30. [PMID: 27688704 PMCID: PMC5024791 DOI: 10.4137/ebo.s40569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 11/05/2022] Open
Abstract
Historically, classifications of arboviruses were based on serological techniques. Hence, collections of arbovirus isolates have been central to this process by providing the antigenic reagents for these methods. However, with increasing concern about biosafety and security, the introduction of molecular biology techniques has led to greater emphasis on the storage of nucleic acid sequence data over the maintenance of archival material. In this commentary, we provide examples of where archival collections provide an important source of genetic material to assist in confirming the authenticity of reference strains and vaccine stocks, to clarify taxonomic relationships particularly when isolates of the same virus species have been collected across a wide expanse of time and space, for future phenotypic analysis, to determine the historical diversity of strains, and to understand the mechanisms leading to changes in genome structure and virus evolution.
Collapse
Affiliation(s)
- Alyssa T Pyke
- Public Health Virology Laboratory, Forensic and Scientific Services, Queensland Health, Archerfield, Queensland, Australia
| | - David Warrilow
- Public Health Virology Laboratory, Forensic and Scientific Services, Queensland Health, Archerfield, Queensland, Australia
| |
Collapse
|
914
|
Simonin Y, Loustalot F, Desmetz C, Foulongne V, Constant O, Fournier-Wirth C, Leon F, Molès JP, Goubaud A, Lemaitre JM, Maquart M, Leparc-Goffart I, Briant L, Nagot N, Van de Perre P, Salinas S. Zika Virus Strains Potentially Display Different Infectious Profiles in Human Neural Cells. EBioMedicine 2016; 12:161-169. [PMID: 27688094 PMCID: PMC5078617 DOI: 10.1016/j.ebiom.2016.09.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 01/28/2023] Open
Abstract
The recent Zika virus (ZIKV) epidemic has highlighted the poor knowledge on its physiopathology. Recent studies showed that ZIKV of the Asian lineage, responsible for this international outbreak, causes neuropathology in vitro and in vivo. However, two African lineages exist and the virus is currently found circulating in Africa. The original African strain was also suggested to be neurovirulent but its laboratory usage has been criticized due to its multiple passages. In this study, we compared the French Polynesian (Asian) ZIKV strain to an African strain isolated in Central African Republic and show a difference in infectivity and cellular response between both strains in human neural stem cells and astrocytes. Consistently, this African strain led to a higher infection rate and viral production, as well as stronger cell death and anti-viral response. Our results highlight the need to better characterize the physiopathology and predict neurological impairment associated with African ZIKV.
Collapse
Affiliation(s)
- Yannick Simonin
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Université de Montpellier, Montpellier, France.
| | - Fabien Loustalot
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | | | - Vincent Foulongne
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Orianne Constant
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | - Chantal Fournier-Wirth
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Etablissement Français du Sang, Montpellier, France
| | - Fanny Leon
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Etablissement Français du Sang, Montpellier, France
| | - Jean-Pierre Molès
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France
| | - Aurélien Goubaud
- Institut de Médecine Régénératrice et Biothérapies, INSERM, U1183, Université de Montpellier, CHU Montpellier, Montpellier, France; Plateforme CHU SAFE-IPS, Infrastructure Nationale INGESTEM, Montpellier, France
| | - Jean-Marc Lemaitre
- Institut de Médecine Régénératrice et Biothérapies, INSERM, U1183, Université de Montpellier, CHU Montpellier, Montpellier, France; Plateforme CHU SAFE-IPS, Infrastructure Nationale INGESTEM, Montpellier, France
| | - Marianne Maquart
- Centre National de Référence des Arbovirus, Institut de Recherche Biomédicale des Armées, Marseille, France
| | - Isabelle Leparc-Goffart
- Centre National de Référence des Arbovirus, Institut de Recherche Biomédicale des Armées, Marseille, France
| | - Laurence Briant
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, FRE3689, CNRS-Université de Montpellier, Montpellier, France
| | - Nicolas Nagot
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Philippe Van de Perre
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France; Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Sara Salinas
- UMR 1058, INSERM, Université de Montpellier, Etablissement Français du Sang Pathogenesis and Control of Chronic Infections, Inserm, Montpellier, France.
| |
Collapse
|
915
|
Ticconi C, Pietropolli A, Rezza G. Zika virus infection and pregnancy: what we do and do not know. Pathog Glob Health 2016; 110:262-268. [PMID: 27690200 DOI: 10.1080/20477724.2016.1234804] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Recent data strongly suggest an association between the current outbreak of ZIKA virus (ZIKV) in many countries of Central and South America and a sharp increase in the detection of microcephaly and fetal malformations. The link with brain defect, which has been detected mainly in some areas of Brazil, is supported by the following evidence: (1) ZIKV transmission from infected pregnant women to their fetuses; (2) the potential of ZIKV to determine a specific congenital fetal syndrome characterized by abnormalities involving primarily the developing brain and eye. In particular, the risk of transmission and congenital disease appears to be restricted to mother's infection during the first trimester of pregnancy. Among brain defects, microcephaly, brain calcifications, and ventriculomegaly are the most frequent abnormalities of the central nervous system detected so far. However, relevant information on effect of maternal infection with ZIKV on the fetus is still limited. In this review, we focus our attention on current knowledge about ZIKV infection in pregnancy, discussing relevant issues and open problems which merit further investigation.
Collapse
Affiliation(s)
- Carlo Ticconi
- a Section of Gynecology and Obstetrics, Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Adalgisa Pietropolli
- a Section of Gynecology and Obstetrics, Department of Biomedicine and Prevention , University of Rome Tor Vergata , Rome , Italy
| | - Giovanni Rezza
- b Department of Infectious Diseases , Istituto Superiore di Sanità , Rome , Italy
| |
Collapse
|
916
|
Silverstein PS, Aljouda NA, Kumar A. Recent developments in vertical transmission of ZIKA virus. Oncotarget 2016; 7:62797-62798. [PMID: 27579616 PMCID: PMC5325328 DOI: 10.18632/oncotarget.11619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/21/2016] [Indexed: 11/29/2022] Open
Affiliation(s)
- Peter S Silverstein
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri, Kansas City, MO, USA
| | - Nour A Aljouda
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri, Kansas City, MO, USA
| | - Anil Kumar
- Division of Pharmacology and Toxicology, School of Pharmacy, University of Missouri, Kansas City, MO, USA
| |
Collapse
|
917
|
Di Guardo G, Braga PBB, Peron JPS. Zika virus-associated brain damage: animal models and open issues. Emerg Microbes Infect 2016; 5:e106. [PMID: 27651092 PMCID: PMC5113054 DOI: 10.1038/emi.2016.103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/15/2016] [Accepted: 07/30/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Giovanni Di Guardo
- Faculty of Veterinary Medicine, University of Teramo, Località Piano d'Accio, Teramo 64100, Italy
| | | | - Jean Pierre Schatzmann Peron
- Department of Immunology, Neuroimmune Interactions Laboratory, University of São Paulo, Saint Paul 05508-000, Brazil
| |
Collapse
|
918
|
Liu S, DeLalio LJ, Isakson BE, Wang TT. AXL-Mediated Productive Infection of Human Endothelial Cells by Zika Virus. Circ Res 2016; 119:1183-1189. [PMID: 27650556 DOI: 10.1161/circresaha.116.309866] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 09/10/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
RATIONALE The mosquito-borne Zika virus (ZIKV) is now recognized as a blood-borne pathogen, raising an important question about how the virus gets into human bloodstream. The imminent threat of the ZIKV epidemic to the global blood supply also demands novel therapeutics to stop virus transmission though transfusion. OBJECTIVE We intend to characterize ZIKV tropism for human endothelial cells (ECs) and provide potential targets for intervention. METHODS AND RESULTS We conducted immunostaining, plaque assay, and quantitative reverse transcription-polymerase chain reaction of ZIKV RNA to evaluate the possible infection of ECs by ZIKV. Both the African and the South American ZIKV strains readily infect human umbilical vein endothelial cells and human ECs derived from aortic and coronary artery, as well as the saphenous vein. Infected ECs released infectious progeny virus. Compared with the African strains, South American ZIKV isolates replicate faster in ECs and are partially cytopathic, suggesting enhanced virulence of these isolates. Flow cytometric analyses showed that the susceptibility of ECs positively correlated with the cell surface levels of tyrosine-protein kinase receptor UFO (AXL) receptor tyrosine kinase. Gain- and loss-of-function studies further revealed that AXL is required for ZIKV entry at a postbinding step. Finally, small-molecule inhibitors of the AXL kinase significantly reduced ZIKA infection of ECs. CONCLUSIONS We identified EC as a key cell type for ZIKV infection. These data support the view of hematogenous dissemination of ZIKV and implicate AXL as a new target for antiviral therapy.
Collapse
Affiliation(s)
- Shufeng Liu
- From the Center for Infectious Diseases, Discovery Biology, SRI International, Harrisonburg, VA (S.L., T.T.W.); and Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (L.J.D., B.E.I.)
| | - Leon J DeLalio
- From the Center for Infectious Diseases, Discovery Biology, SRI International, Harrisonburg, VA (S.L., T.T.W.); and Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (L.J.D., B.E.I.)
| | - Brant E Isakson
- From the Center for Infectious Diseases, Discovery Biology, SRI International, Harrisonburg, VA (S.L., T.T.W.); and Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (L.J.D., B.E.I.)
| | - Tony T Wang
- From the Center for Infectious Diseases, Discovery Biology, SRI International, Harrisonburg, VA (S.L., T.T.W.); and Department of Molecular Physiology and Biophysics, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (L.J.D., B.E.I.).
| |
Collapse
|
919
|
Alvarado MG, Schwartz DA. Zika Virus Infection in Pregnancy, Microcephaly, and Maternal and Fetal Health: What We Think, What We Know, and What We Think We Know. Arch Pathol Lab Med 2016; 141:26-32. [PMID: 27636525 DOI: 10.5858/arpa.2016-0382-ra] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT -The global epidemic of Zika virus (ZIKV) infection has emerged as an important public health problem affecting pregnant women and their infants. OBJECTIVES -To review the causal association between ZIKV infection during pregnancy and intrauterine fetal infection, microcephaly, brain damage, congenital malformation syndrome, and experimental laboratory models of fetal infection. Many questions remain regarding the risk factors, pathophysiology, epidemiology, and timing of maternal-fetal transmission and disease. These include mechanisms of fetal brain damage and microcephaly; the role of covariables, such as viral burden, duration of viremia, and host genetics, on vertical transmission; and the clinical and pathologic spectrum of congenital Zika syndrome. Additional questions include defining the potential long-term physical and neurobehavioral outcomes for infected infants, whether maternal or fetal host genetics influence the clinical outcome, and whether ZIKV infection can cause maternal morbidity. Finally, are experimental laboratory and animal models of ZIKV infection helpful in addressing maternal-fetal viral transmission and the development of congenital microcephaly? This communication provides current information and attempts to address some of these important questions. DATA SOURCES -Comprehensive review of published scientific literature. CONCLUSIONS -Recent advances in epidemiology, clinical medicine, pathology, and experimental studies have provided a great amount of new information regarding vertical ZIKV transmission and the mechanisms of congenital microcephaly, brain damage, and congenital Zika syndrome in a relatively short time. However, much work still needs to be performed to more completely understand the maternal and fetal aspects of this new and emerging viral disease.
Collapse
Affiliation(s)
| | - David A Schwartz
- From the Department of Anthropology, Georgia State University, Atlanta (Dr Alvarado); and the Department of Pathology, Medical College of Georgia, Augusta University, Augusta (Dr Schwartz)
| |
Collapse
|
920
|
Abbink P, Larocca RA, De La Barrera RA, Bricault CA, Moseley ET, Boyd M, Kirilova M, Li Z, Ng'ang'a D, Nanayakkara O, Nityanandam R, Mercado NB, Borducchi EN, Agarwal A, Brinkman AL, Cabral C, Chandrashekar A, Giglio PB, Jetton D, Jimenez J, Lee BC, Mojta S, Molloy K, Shetty M, Neubauer GH, Stephenson KE, Peron JPS, Zanotto PMDA, Misamore J, Finneyfrock B, Lewis MG, Alter G, Modjarrad K, Jarman RG, Eckels KH, Michael NL, Thomas SJ, Barouch DH. Protective efficacy of multiple vaccine platforms against Zika virus challenge in rhesus monkeys. Science 2016; 353:1129-32. [PMID: 27492477 PMCID: PMC5237380 DOI: 10.1126/science.aah6157] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 01/10/2023]
Abstract
Zika virus (ZIKV) is responsible for a major ongoing epidemic in the Americas and has been causally associated with fetal microcephaly. The development of a safe and effective ZIKV vaccine is therefore an urgent global health priority. Here we demonstrate that three different vaccine platforms protect against ZIKV challenge in rhesus monkeys. A purified inactivated virus vaccine induced ZIKV-specific neutralizing antibodies and completely protected monkeys against ZIKV strains from both Brazil and Puerto Rico. Purified immunoglobulin from vaccinated monkeys also conferred passive protection in adoptive transfer studies. A plasmid DNA vaccine and a single-shot recombinant rhesus adenovirus serotype 52 vector vaccine, both expressing ZIKV premembrane and envelope, also elicited neutralizing antibodies and completely protected monkeys against ZIKV challenge. These data support the rapid clinical development of ZIKV vaccines for humans.
Collapse
Affiliation(s)
- Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Christine A Bricault
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Edward T Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Boyd
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Marinela Kirilova
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhenfeng Li
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Ng'ang'a
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ovini Nanayakkara
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramya Nityanandam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Noe B Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Arshi Agarwal
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Amanda L Brinkman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Crystal Cabral
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Patricia B Giglio
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Jetton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jessica Jimenez
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Benjamin C Lee
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Shanell Mojta
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Katherine Molloy
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mayuri Shetty
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - George H Neubauer
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kathryn E Stephenson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | | | | | | | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Kayvon Modjarrad
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA. Henry M. Jackson Foundation, Bethesda, MD 20817, USA
| | - Richard G Jarman
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Kenneth H Eckels
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Nelson L Michael
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Stephen J Thomas
- Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
921
|
Yin Y, Xu Y, Su L, Zhu X, Chen M, Zhu W, Xia H, Huang X, Gong S. Epidemiologic investigation of a family cluster of imported ZIKV cases in Guangdong, China: probable human-to-human transmission. Emerg Microbes Infect 2016; 5:e100. [PMID: 27599469 PMCID: PMC5113051 DOI: 10.1038/emi.2016.100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 07/22/2016] [Accepted: 08/01/2016] [Indexed: 12/12/2022]
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that can potentially threaten South China. A Chinese family of four returning from Venezuela to China was found to be positive for ZIKV when the youngest son's fever was first detected at an airport immigration inspection. They were isolated temporarily in a local hospital in Enping city, Guangdong province, where their clinical data were recorded and urine and saliva were collected to isolate ZIKV and to obtain viral sequences. All of them except the mother presented mild symptoms of rash and fever. Envelope gene sequences from the father, daughter and son were completely identical. Phylogenetic analysis demonstrated that this strain is similar to several imported strains reported in recent months, which are all clustered into a group isolated from 2015 ZIKA outbreaks in Brazil. Together with the climatic features in Venezuela, New York and Guangdong in February, it can be concluded that our subjects are imported cases from Venezuela. With the same viral sequence being shared between family members, neither direct human-to-human nor vector transmission can be ruled out in this study, but the former seems more likely. Although our subjects had mild illness, epidemiologists and public health officials should be aware of the risk of further expansion of ZIKV transmission by local competent vectors.
Collapse
Affiliation(s)
- Yingxian Yin
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yi Xu
- Department of Infectious Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ling Su
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xun Zhu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Minxia Chen
- Department of Infectious Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Weijin Zhu
- Department of Infectious Diseases, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xi Huang
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| |
Collapse
|
922
|
Onorati M, Li Z, Liu F, Sousa AMM, Nakagawa N, Li M, Dell'Anno MT, Gulden FO, Pochareddy S, Tebbenkamp ATN, Han W, Pletikos M, Gao T, Zhu Y, Bichsel C, Varela L, Szigeti-Buck K, Lisgo S, Zhang Y, Testen A, Gao XB, Mlakar J, Popovic M, Flamand M, Strittmatter SM, Kaczmarek LK, Anton ES, Horvath TL, Lindenbach BD, Sestan N. Zika Virus Disrupts Phospho-TBK1 Localization and Mitosis in Human Neuroepithelial Stem Cells and Radial Glia. Cell Rep 2016; 16:2576-2592. [PMID: 27568284 PMCID: PMC5135012 DOI: 10.1016/j.celrep.2016.08.038] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/27/2016] [Accepted: 08/12/2016] [Indexed: 02/08/2023] Open
Abstract
The mechanisms underlying Zika virus (ZIKV)-related microcephaly and other neurodevelopment defects remain poorly understood. Here, we describe the derivation and characterization, including single-cell RNA-seq, of neocortical and spinal cord neuroepithelial stem (NES) cells to model early human neurodevelopment and ZIKV-related neuropathogenesis. By analyzing human NES cells, organotypic fetal brain slices, and a ZIKV-infected micrencephalic brain, we show that ZIKV infects both neocortical and spinal NES cells as well as their fetal homolog, radial glial cells (RGCs), causing disrupted mitoses, supernumerary centrosomes, structural disorganization, and cell death. ZIKV infection of NES cells and RGCs causes centrosomal depletion and mitochondrial sequestration of phospho-TBK1 during mitosis. We also found that nucleoside analogs inhibit ZIKV replication in NES cells, protecting them from ZIKV-induced pTBK1 relocalization and cell death. We established a model system of human neural stem cells to reveal cellular and molecular mechanisms underlying neurodevelopmental defects associated with ZIKV infection and its potential treatment.
Collapse
Affiliation(s)
- Marco Onorati
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Zhen Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Fuchen Liu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - André M M Sousa
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Naoki Nakagawa
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Mingfeng Li
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Maria Teresa Dell'Anno
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Forrest O Gulden
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Sirisha Pochareddy
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew T N Tebbenkamp
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Wenqi Han
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mihovil Pletikos
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Tianliuyun Gao
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Ying Zhu
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Candace Bichsel
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Luis Varela
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Klara Szigeti-Buck
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Steven Lisgo
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE13BZ, UK
| | - Yalan Zhang
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anze Testen
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Xiao-Bing Gao
- Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA
| | - Jernej Mlakar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Mara Popovic
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana 1000, Slovenia
| | - Marie Flamand
- Department of Virology, Institut Pasteur, 75724 Paris Cedex 15, France
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Leonard K Kaczmarek
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06510, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06510, USA
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Tamas L Horvath
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA; Yale Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Brett D Lindenbach
- Department of Microbial Pathogenesis, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Nenad Sestan
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Cellular Neuroscience, Neurodegeneration and Repair Program, Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Section of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Genetics and Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
923
|
Miner JJ, Sene A, Richner JM, Smith AM, Santeford A, Ban N, Weger-Lucarelli J, Manzella F, Rückert C, Govero J, Noguchi KK, Ebel GD, Diamond MS, Apte RS. Zika Virus Infection in Mice Causes Panuveitis with Shedding of Virus in Tears. Cell Rep 2016; 16:3208-3218. [PMID: 27612415 DOI: 10.1016/j.celrep.2016.08.079] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 08/12/2016] [Accepted: 08/25/2016] [Indexed: 12/12/2022] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital abnormalities and Guillain-Barré syndrome. ZIKV infection also results in severe eye disease characterized by optic neuritis, chorioretinal atrophy, and blindness in newborns and conjunctivitis and uveitis in adults. We evaluated ZIKV infection of the eye by using recently developed mouse models of pathogenesis. ZIKV-inoculated mice developed conjunctivitis, panuveitis, and infection of the cornea, iris, optic nerve, and ganglion and bipolar cells in the retina. This phenotype was independent of the entry receptors Axl or Mertk, given that Axl(-/-), Mertk(-/-), and Axl(-/-)Mertk(-/-) double knockout mice sustained levels of infection similar to those of control animals. We also detected abundant viral RNA in tears, suggesting that virus might be secreted from lacrimal glands or shed from the cornea. This model provides a foundation for studying ZIKV-induced ocular disease, defining mechanisms of viral persistence, and developing therapeutic approaches for viral infections of the eye.
Collapse
Affiliation(s)
- Jonathan J Miner
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US.
| | - Abdoulaye Sene
- Department of Ophthalmology and Visual Sciences, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - Justin M Richner
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - Amber M Smith
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - Andrea Santeford
- Department of Ophthalmology and Visual Sciences, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - Norimitsu Ban
- Department of Ophthalmology and Visual Sciences, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - James Weger-Lucarelli
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Francesca Manzella
- Department of Psychiatry, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Claudia Rückert
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Jennifer Govero
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US
| | - Kevin K Noguchi
- Department of Psychiatry, School of Medicine, Washington University, Saint Louis, MO 63110, USA
| | - Gregory D Ebel
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Michael S Diamond
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US; Department of Molecular Microbiology, School of Medicine, Washington University, Saint Louis, MO 63110, USA; Department of Pathology and Immunology, School of Medicine, Washington University, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, School of Medicine, Washington University, Saint Louis, MO 63110, USA.
| | - Rajendra S Apte
- Department of Medicine, School of Medicine, Washington University, Saint Louis, MO 63110, US; Department of Ophthalmology and Visual Sciences, School of Medicine, Washington University, Saint Louis, MO 63110, US; Department of Developmental Biology, School of Medicine, Washington University, Saint Louis, MO 63110, USA.
| |
Collapse
|
924
|
Paes de Andrade P, Aragão FJL, Colli W, Dellagostin OA, Finardi-Filho F, Hirata MH, Lira-Neto ADC, Almeida de Melo M, Nepomuceno AL, Gorgônio da Nóbrega F, Delfino de Sousa G, Valicente FH, Zanettini MHB. Use of transgenic Aedes aegypti in Brazil: risk perception and assessment. Bull World Health Organ 2016; 94:766-771. [PMID: 27843167 PMCID: PMC5043214 DOI: 10.2471/blt.16.173377] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 06/16/2016] [Accepted: 06/22/2016] [Indexed: 01/29/2023] Open
Abstract
The OX513A strain of Aedes aegypti, which was developed by the British company Oxitec, expresses a self-limiting transgene that prevents larvae from developing to adulthood. In April 2014, the Brazilian National Technical Commission on Biosafety completed a risk assessment of OX513A and concluded that the strain did not present new biological risks to humans or the environment and could be released in Brazil. At that point, Brazil became the first country to approve the unconstrained release of a genetically modified mosquito. During the assessment, the commission produced a comprehensive list of – and systematically analysed – the perceived hazards. Such hazards included the potential survival to adulthood of immature stages carrying the transgene – should the transgene fail to be expressed or be turned off by exposure to sufficient environmental tetracycline. Other perceived hazards included the potential allergenicity and/or toxicity of the proteins expressed by the gene, the potential for gene flow or increased transmission of human pathogens and the occupation of vacant breeding sites by other vector species. The Zika epidemic both elevated the perceived importance of Ae. aegypti as a vector – among policy-makers and regulators as well as the general public – and increased concerns over the release of males of the OX513A strain. We have therefore reassessed the potential hazards. We found that release of the transgenic mosquitoes would still be both safe and of great potential value in the control of diseases spread by Ae. aegypti, such as chikungunya, dengue and Zika.
Collapse
Affiliation(s)
- Paulo Paes de Andrade
- Departamento de Genética, Universidade Federal de Pernambuco, Avenida Moraes Rego s/s, 50670-901, Recife, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
925
|
|
926
|
Affiliation(s)
- Jörg Heukelbach
- Department of Community Health, School of Medicine, Federal University of Ceará, Rodolfo Teófilo, 60430-140 Fortaleza, CE, Brazil; Anton Breinl Centre for Public Health and College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University, Townsville, QLD, Australia.
| | - Guilherme Loureiro Werneck
- Department of Epidemiology, Social Medicine Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
927
|
Larocca RA, Abbink P, Peron JPS, de A. Zanotto PM, Iampietro MJ, Badamchi-Zadeh A, Boyd M, Ng’ang’a D, Kirilova M, Nityanandam R, Mercado NB, Li Z, Moseley ET, Bricault CA, Borducchi EN, Giglio PB, Jetton D, Neubauer G, Nkolola JP, Maxfield LF, De La Barrera RA, Jarman RG, Eckels KH, Michael NL, Thomas SJ, Barouch DH. Vaccine protection against Zika virus from Brazil. Nature 2016; 536:474-8. [PMID: 27355570 PMCID: PMC5003703 DOI: 10.1038/nature18952] [Citation(s) in RCA: 406] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 06/22/2016] [Indexed: 12/23/2022]
Abstract
Zika virus (ZIKV) is a flavivirus that is responsible for the current epidemic in Brazil and the Americas. ZIKV has been causally associated with fetal microcephaly, intrauterine growth restriction, and other birth defects in both humans and mice. The rapid development of a safe and effective ZIKV vaccine is a global health priority, but very little is currently known about ZIKV immunology and mechanisms of immune protection. Here we show that a single immunization with a plasmid DNA vaccine or a purified inactivated virus vaccine provides complete protection in susceptible mice against challenge with a strain of ZIKV involved in the outbreak in northeast Brazil. This ZIKV strain has recently been shown to cross the placenta and to induce fetal microcephaly and other congenital malformations in mice. We produced DNA vaccines expressing ZIKV pre-membrane and envelope (prM-Env), as well as a series of deletion mutants. The prM-Env DNA vaccine, but not the deletion mutants, afforded complete protection against ZIKV, as measured by absence of detectable viraemia following challenge, and protective efficacy correlated with Env-specific antibody titers. Adoptive transfer of purified IgG from vaccinated mice conferred passive protection, and depletion of CD4 and CD8 T lymphocytes in vaccinated mice did not abrogate this protection. These data demonstrate that protection against ZIKV challenge can be achieved by single-shot subunit and inactivated virus vaccines in mice and that Env-specific antibody titers represent key immunologic correlates of protection. Our findings suggest that the development of a ZIKV vaccine for humans is likely to be achievable.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Antibody Specificity
- Brazil
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Deletion
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/isolation & purification
- Mice
- Microcephaly/complications
- Microcephaly/virology
- Vaccines, DNA/chemistry
- Vaccines, DNA/genetics
- Vaccines, DNA/immunology
- Vaccines, Inactivated/chemistry
- Vaccines, Inactivated/genetics
- Vaccines, Inactivated/immunology
- Vaccines, Subunit/chemistry
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
- Viral Vaccines/chemistry
- Viral Vaccines/genetics
- Viral Vaccines/immunology
- Zika Virus/chemistry
- Zika Virus/genetics
- Zika Virus/immunology
- Zika Virus Infection/complications
- Zika Virus Infection/immunology
- Zika Virus Infection/prevention & control
- Zika Virus Infection/virology
Collapse
Affiliation(s)
- Rafael A. Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | - M. Justin Iampietro
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alexander Badamchi-Zadeh
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Michael Boyd
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Ng’ang’a
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Marinela Kirilova
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ramya Nityanandam
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Noe B. Mercado
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhenfeng Li
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Edward T. Moseley
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christine A. Bricault
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Erica N. Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Patricia B. Giglio
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David Jetton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - George Neubauer
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph P. Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lori F. Maxfield
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | - Richard G. Jarman
- Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | - Kenneth H. Eckels
- Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | - Nelson L. Michael
- Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | - Stephen J. Thomas
- Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
928
|
Vouga M, Baud D. Imaging of congenital Zika virus infection: the route to identification of prognostic factors. Prenat Diagn 2016; 36:799-811. [PMID: 27481629 DOI: 10.1002/pd.4880] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 07/24/2016] [Accepted: 07/25/2016] [Indexed: 12/16/2022]
Abstract
Zika virus (ZIKV) has recently emerged as a novel teratogenic agent associated with severe neurological complications. The risk associated with maternal infection remains to be exactly defined but appears to be significant. Like other TORCH agents (toxoplasmosis, other agents, rubella, cytomegalovirus and herpes simplex), it is unlikely that all affected fetuses will be symptomatic at birth. It is therefore urgent to better define the spectrum of anomalies observed in infected fetuses to provide adequate parental counseling. In this review, we provide a comprehensive analysis of major cases described to date and highlight specific prenatal and postnatal radiological findings of congenital ZIKV infection. A total of 19 reports were included in our analysis. ZIKV seemed to harbor a specific tropism for the central nervous system, and anomalies were mostly limited to the brain. Major radiological findings were ventriculomegaly, diffuse calcifications and signs of abnormal gyration as well as cortical development. In addition, a significant number of fetuses suffered from intra uterine growth restriction. Based on these findings, we provide recommendations for adequate radiological monitoring of at-risk pregnancies. © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Manon Vouga
- Materno-fetal and Obstetrics Research Unit, Department of Obstetrics and Gynecology, Maternity, University Hospital, Lausanne, Switzerland
| | - David Baud
- Institute of Microbiology, Faculty of Biology and Medicine, University of Lausanne and University Hospital, Lausanne, Switzerland.
| |
Collapse
|
929
|
Schwartz DA. Autopsy and Postmortem Studies Are Concordant: Pathology of Zika Virus Infection Is Neurotropic in Fetuses and Infants With Microcephaly Following Transplacental Transmission. Arch Pathol Lab Med 2016; 141:68-72. [PMID: 27557413 DOI: 10.5858/arpa.2016-0343-oa] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT -Pathology studies have been important in concluding that Zika virus infection occurring in pregnant women can result in vertical transmission of the agent from mother to fetus. Fetal and infant autopsies have provided crucial direct evidence that Zika virus can infect an unborn child, resulting in microcephaly, other malformations, and, in some cases, death. OBJECTIVE -To better understand the etiologic role and mechanism(s) of Zika virus in causing birth defects such as microcephaly, this communication analyzes the spectrum of clinical and autopsy studies reported from fetuses and infants who developed intrauterine Zika virus infection, and compares these findings with experimental data related to Zika virus infection. DESIGN -Retrospective analysis of reported clinical, autopsy, pathology, and related postmortem studies from 9 fetuses and infants with intrauterine Zika virus infection and microcephaly. RESULTS -All fetuses and infants examined demonstrated an overlapping spectrum of gross and microscopic neuropathologic abnormalities. Direct cytopathic effects of infection by the Zika virus were confined to the brain; in cases where other organs were evaluated, no direct viral effects were identified. CONCLUSIONS -There is concordance of the spectrum of brain damage, reinforcing previous data indicating that the Zika virus has a strong predilection for cells of the fetal central nervous system following vertical transmission. The occurrence of additional congenital abnormalities suggests that intrauterine brain damage from Zika virus interferes with normal fetal development, resulting in fetal akinesia. Experimental in vitro and in vivo studies of Zika virus infection corroborate the human autopsy findings of neural specificity.
Collapse
Affiliation(s)
- David A Schwartz
- From the Department of Pathology, Medical College of Georgia, Augusta University, Augusta
| |
Collapse
|
930
|
Barzon L, Trevisan M, Sinigaglia A, Lavezzo E, Palù G. Zika virus: from pathogenesis to disease control. FEMS Microbiol Lett 2016; 363:fnw202. [PMID: 27549304 DOI: 10.1093/femsle/fnw202] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2016] [Indexed: 12/21/2022] Open
Abstract
Zika virus is a mosquito-borne flavivirus discovered in Uganda in 1947. The virus has emerged in recent years and spread in the Pacific Area and the Americas, where it has caused large human outbreaks. The factors involved in the virus's emergence are still unknown, but probably include its introduction in naïve environments characterised by the presence of high densities of competent Aedes spp. mosquitoes and susceptible human hosts in urban areas. Unique features of Zika virus infection are sexual and transplacental transmission and associated neurological morbidities, i.e. Guillain-Barré syndrome and fetal microcephaly. Diagnosis relies on the detection of viral nucleic acids in biological samples, while detection of a specific antibody response may be inconclusive because of the broad cross-reactivity of antibodies among flaviviruses. Experimental studies have clarified some mechanisms of Zika virus pathogenesis and have identified potential targets for antiviral drugs. In animal models, the virus can infect and efficiently replicate in the placenta and in the brain, and induce fetal demise or neural damage, recapitulating human diseases. These animal models have been used to evaluate candidate vaccines and promising results have been obtained.
Collapse
Affiliation(s)
- Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marta Trevisan
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Enrico Lavezzo
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Padova, Italy
| |
Collapse
|
931
|
Li H, Saucedo-Cuevas L, Regla-Nava JA, Chai G, Sheets N, Tang W, Terskikh AV, Shresta S, Gleeson JG. Zika Virus Infects Neural Progenitors in the Adult Mouse Brain and Alters Proliferation. Cell Stem Cell 2016; 19:593-598. [PMID: 27545505 PMCID: PMC5097023 DOI: 10.1016/j.stem.2016.08.005] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/18/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022]
Abstract
Zika virus (ZIKV)-related neuropathology is an important global health concern. Several studies have shown that ZIKV can infect neural stem cells in the developing brain, but infection in the adult brain has not been examined. Two areas in the adult mouse brain contain neural stem cells: the subventricular zone of the anterior forebrain and the subgranular zone of the hippocampus. Here, using 6-week-old mice triply deficient in interferon regulatory factor (IRF) as a model, we show that blood-borne ZIKV administration can lead to pronounced evidence of ZIKV infection in these adult neural stem cells, leading to cell death and reduced proliferation. Our data therefore suggest that adult as well as fetal neural stem cells are vulnerable to ZIKV neuropathology. Thus, although ZIKV is considered a transient infection in adult humans without marked long-term effects, there may in fact be consequences of exposure in the adult brain.
Collapse
Affiliation(s)
- Hongda Li
- Laboratory for Pediatric Brain Diseases, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA 10065 and Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Laura Saucedo-Cuevas
- Laboratory for Pediatric Brain Diseases, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA 10065 and Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Jose A Regla-Nava
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA 92037
| | - Guoliang Chai
- Laboratory for Pediatric Brain Diseases, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA 10065 and Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Nicholas Sheets
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA 92037
| | - William Tang
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA 92037
| | - Alexey V Terskikh
- Del. E. Webb Center for Neuroscience, Aging and Stem Cell Regeneration, Sanford Burnham Prebys Discovery Institute, La Jolla, CA, USA 92037
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy & Immunology, La Jolla, CA, USA 92037
| | - Joseph G Gleeson
- Laboratory for Pediatric Brain Diseases, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA 10065 and Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
932
|
Jurado KA, Simoni MK, Tang Z, Uraki R, Hwang J, Householder S, Wu M, Lindenbach BD, Abrahams VM, Guller S, Fikrig E. Zika virus productively infects primary human placenta-specific macrophages. JCI Insight 2016; 1. [PMID: 27595140 DOI: 10.1172/jci.insight.88461] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The strong association of Zika virus infection with congenital defects has led to questions of how a flavivirus is capable of crossing the placental barrier to reach the fetal brain. Here, we demonstrate permissive Zika virus infection of primary human placental macrophages, commonly referred to as Hofbauer cells, and placental villous fibroblasts. We also demonstrate Zika virus infection of Hofbauer cells within the context of the tissue ex vivo using term placental villous explants. In addition to amplifying infectious virus within a usually inaccessible area, the putative migratory activities of Hofbauer cells may aid in dissemination of Zika virus to the fetal brain. Understanding the susceptibility of placenta-specific cell types will aid future work around and understanding of Zika virus-associated pregnancy complications.
Collapse
Affiliation(s)
- Kellie Ann Jurado
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Michael K Simoni
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ryuta Uraki
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jesse Hwang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sarah Householder
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Mingjie Wu
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Brett D Lindenbach
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| |
Collapse
|
933
|
Lessler J, Chaisson LH, Kucirka LM, Bi Q, Grantz K, Salje H, Carcelen AC, Ott CT, Sheffield JS, Ferguson NM, Cummings DAT, Metcalf CJE, Rodriguez-Barraquer I. Assessing the global threat from Zika virus. Science 2016; 353:aaf8160. [PMID: 27417495 PMCID: PMC5467639 DOI: 10.1126/science.aaf8160] [Citation(s) in RCA: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
First discovered in 1947, Zika virus (ZIKV) infection remained a little-known tropical disease until 2015, when its apparent association with a considerable increase in the incidence of microcephaly in Brazil raised alarms worldwide. There is limited information on the key factors that determine the extent of the global threat from ZIKV infection and resulting complications. Here, we review what is known about the epidemiology, natural history, and public health effects of ZIKV infection, the empirical basis for this knowledge, and the critical knowledge gaps that need to be filled.
Collapse
Affiliation(s)
- Justin Lessler
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | - Lelia H Chaisson
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lauren M Kucirka
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qifang Bi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kyra Grantz
- Department of Biology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - Henrik Salje
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Paris, France
| | - Andrea C Carcelen
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Cassandra T Ott
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jeanne S Sheffield
- Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Neil M Ferguson
- Department of Medicine, School of Public Health, Imperial College London, London, UK
| | - Derek A T Cummings
- Department of Biology, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - C Jessica E Metcalf
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA. Office of Population Research, Princeton University, Princeton, NJ, USA
| | | |
Collapse
|
934
|
Zhao H, Fernandez E, Dowd KA, Speer SD, Platt DJ, Gorman MJ, Govero J, Nelson CA, Pierson TC, Diamond MS, Fremont DH. Structural Basis of Zika Virus-Specific Antibody Protection. Cell 2016; 166:1016-1027. [PMID: 27475895 PMCID: PMC4983199 DOI: 10.1016/j.cell.2016.07.020] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/14/2016] [Accepted: 07/14/2016] [Indexed: 11/18/2022]
Abstract
Zika virus (ZIKV) infection during pregnancy has emerged as a global public health problem because of its ability to cause severe congenital disease. Here, we developed six mouse monoclonal antibodies (mAbs) against ZIKV including four (ZV-48, ZV-54, ZV-64, and ZV-67) that were ZIKV specific and neutralized infection of African, Asian, and American strains to varying degrees. X-ray crystallographic and competition binding analyses of Fab fragments and scFvs defined three spatially distinct epitopes in DIII of the envelope protein corresponding to the lateral ridge (ZV-54 and ZV-67), C-C' loop (ZV-48 and ZV-64), and ABDE sheet (ZV-2) regions. In vivo passive transfer studies revealed protective activity of DIII-lateral ridge specific neutralizing mAbs in a mouse model of ZIKV infection. Our results suggest that DIII is targeted by multiple type-specific antibodies with distinct neutralizing activity, which provides a path for developing prophylactic antibodies for use in pregnancy or designing epitope-specific vaccines against ZIKV.
Collapse
Affiliation(s)
- Haiyan Zhao
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Estefania Fernandez
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott D Speer
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Derek J Platt
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Matthew J Gorman
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Christopher A Nelson
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael S Diamond
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA; Department of Biochemistry & Molecular Biophysics, Washington University School of Medicine, Saint Louis, MO 63110, USA; The Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| |
Collapse
|
935
|
Liang Q, Luo Z, Zeng J, Chen W, Foo SS, Lee SA, Ge J, Wang S, Goldman SA, Zlokovic BV, Zhao Z, Jung JU. Zika Virus NS4A and NS4B Proteins Deregulate Akt-mTOR Signaling in Human Fetal Neural Stem Cells to Inhibit Neurogenesis and Induce Autophagy. Cell Stem Cell 2016; 19:663-671. [PMID: 27524440 DOI: 10.1016/j.stem.2016.07.019] [Citation(s) in RCA: 383] [Impact Index Per Article: 47.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 06/20/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022]
Abstract
The current widespread outbreak of Zika virus (ZIKV) infection has been linked to severe clinical birth defects, particularly microcephaly, warranting urgent study of the molecular mechanisms underlying ZIKV pathogenesis. Akt-mTOR signaling is one of the key cellular pathways essential for brain development and autophagy regulation. Here, we show that ZIKV infection of human fetal neural stem cells (fNSCs) causes inhibition of the Akt-mTOR pathway, leading to defective neurogenesis and aberrant activation of autophagy. By screening the three structural proteins and seven nonstructural proteins present in ZIKV, we found that two, NS4A and NS4B, cooperatively suppress the Akt-mTOR pathway and lead to cellular dysregulation. Corresponding proteins from the closely related dengue virus do not have the same effect on neurogenesis. Thus, our study highlights ZIKV NS4A and NS4B as candidate determinants of viral pathogenesis and identifies a mechanism of action for their effects, suggesting potential targets for anti-ZIKV therapeutic intervention.
Collapse
Affiliation(s)
- Qiming Liang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhifei Luo
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jianxiong Zeng
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Weiqiang Chen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Suan-Sin Foo
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shin-Ae Lee
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jianning Ge
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Su Wang
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14642, USA; Department of Neurology, University of Rochester, Rochester, NY 14642, USA; Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester, Rochester, NY 14642, USA; Department of Neurology, University of Rochester, Rochester, NY 14642, USA; Faculty of Health and Medical Sciences, University of Copenhagen, 1165 Copenhagen, Denmark
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Zhen Zhao
- Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Jae U Jung
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
936
|
Zika virus disease: a current review of the literature. Infection 2016; 44:695-705. [DOI: 10.1007/s15010-016-0935-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/02/2016] [Indexed: 10/21/2022]
|
937
|
Barrows NJ, Campos RK, Powell ST, Prasanth KR, Schott-Lerner G, Soto-Acosta R, Galarza-Muñoz G, McGrath EL, Urrabaz-Garza R, Gao J, Wu P, Menon R, Saade G, Fernandez-Salas I, Rossi SL, Vasilakis N, Routh A, Bradrick SS, Garcia-Blanco MA. A Screen of FDA-Approved Drugs for Inhibitors of Zika Virus Infection. Cell Host Microbe 2016; 20:259-70. [PMID: 27476412 PMCID: PMC4993926 DOI: 10.1016/j.chom.2016.07.004] [Citation(s) in RCA: 365] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 06/19/2016] [Accepted: 07/06/2016] [Indexed: 12/13/2022]
Abstract
Currently there are no approved vaccines or specific therapies to prevent or treat Zika virus (ZIKV) infection. We interrogated a library of FDA-approved drugs for their ability to block infection of human HuH-7 cells by a newly isolated ZIKV strain (ZIKV MEX_I_7). More than 20 out of 774 tested compounds decreased ZIKV infection in our in vitro screening assay. Selected compounds were further validated for inhibition of ZIKV infection in human cervical, placental, and neural stem cell lines, as well as primary human amnion cells. Established anti-flaviviral drugs (e.g., bortezomib and mycophenolic acid) and others that had no previously known antiviral activity (e.g., daptomycin) were identified as inhibitors of ZIKV infection. Several drugs reduced ZIKV infection across multiple cell types. This study identifies drugs that could be tested in clinical studies of ZIKV infection and provides a resource of small molecules to study ZIKV pathogenesis.
Collapse
Affiliation(s)
- Nicholas J Barrows
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Rafael K Campos
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27710, USA
| | - Steven T Powell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA
| | - K Reddisiva Prasanth
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA
| | - Geraldine Schott-Lerner
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA
| | - Ruben Soto-Acosta
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA
| | - Gaddiel Galarza-Muñoz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA
| | - Erica L McGrath
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Rheanna Urrabaz-Garza
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Junling Gao
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Ping Wu
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - George Saade
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Ildefonso Fernandez-Salas
- Centro Regional de Investigación en Salud Publica INSP, 19 Poniente Esquina 4(a) Norte s/n, Colonia Centro, Tapachula, Chiapas 30700, C.P., Mexico
| | - Shannan L Rossi
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, and Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Nikos Vasilakis
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases, and Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Andrew Routh
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA; Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA.
| | - Mariano A Garcia-Blanco
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, 301 University Blvd, Galveston, TX 77555, USA; Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore 169857, Republic of Singapore.
| |
Collapse
|
938
|
Mason JO, Price DJ. Building brains in a dish: Prospects for growing cerebral organoids from stem cells. Neuroscience 2016; 334:105-118. [PMID: 27506142 DOI: 10.1016/j.neuroscience.2016.07.048] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 07/22/2016] [Accepted: 07/30/2016] [Indexed: 11/27/2022]
Abstract
The recent development of organoid techniques, in which embryonic brain-like tissue can be grown from human or mouse stem cells in vitro offers the potential to transform the way in which brain development is studied. In this review, we summarize key aspects of the embryonic development of mammalian forebrains, focussing in particular on the cerebral cortex and highlight significant differences between mouse and primates, including human. We discuss recent work using cerebral organoids that has revealed key similarities and differences between their development and that of the brain in vivo. Finally, we outline the ways in which cerebral organoids can be used in combination with CRISPR/Cas9 genome editing to unravel genetic mechanisms that control embryonic development of the cerebral cortex, how this can help us understand the causes of neurodevelopmental disorders and some of the key challenges which will have to be resolved before organoids can become a mainstream tool to study brain development.
Collapse
Affiliation(s)
- John O Mason
- Centre for Integrative Physiology, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| | - David J Price
- Centre for Integrative Physiology, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK.
| |
Collapse
|
939
|
|
940
|
Affiliation(s)
- Indira U Mysorekar
- From the Departments of Pathology and Immunology (I.U.M., M.S.D.), Obstetrics and Gynecology (I.U.M.), and Medicine and Molecular Microbiology (M.S.D.), and the Center for Human Immunology and Immunotherapy Programs (M.S.D.), Washington University School of Medicine, St. Louis
| | - Michael S Diamond
- From the Departments of Pathology and Immunology (I.U.M., M.S.D.), Obstetrics and Gynecology (I.U.M.), and Medicine and Molecular Microbiology (M.S.D.), and the Center for Human Immunology and Immunotherapy Programs (M.S.D.), Washington University School of Medicine, St. Louis
| |
Collapse
|
941
|
Abstract
The epidemics of Ebola virus in West Africa and Zika virus in America highlight how viruses can explosively emerge into new territories. These epidemics also exposed how unprepared we are to handle infectious disease emergencies. This is also true when we consider hypothesized new clinical features of infection, such as the associations between Zika virus infection and severe neurological disease, including microcephaly and Guillain-Barré syndrome. On the surface, these pathologies appear to be new features of Zika virus infection, however, causal relationships have not yet been established. Decades of limited Zika virus research are making us scramble to determine the true drivers behind the epidemic, often at the expense of over-speculation without credible evidence. Here we review the literature and find no conclusive evidence at this time for significant biological differences between the American Zika virus strains and those circulating elsewhere. Rather, the epidemic scale in the Americas may be facilitated by an abnormally warm climate, dense human and mosquito populations, and previous exposure to other viruses. Severe disease associated with Zika virus may therefore not be a new trait for the virus, rather it may have been overlooked due to previously small outbreaks. Much of the recent panic regarding Zika virus has been about the Olympics in Brazil. We do not find any substantial evidence that the Olympics will result in a significant number of new Zika virus infections (~10 predicted) or that the Olympics will promote further epidemic spread over what is already expected. The Zika virus epidemic in the Americas is a serious situation and decisions based on solid scientific evidence - not hyped media speculations - are required for effective outbreak response.
Collapse
Affiliation(s)
- Nathan D. Grubaugh
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Kristian G. Andersen
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Scripps Translational Science Institute, La Jolla, CA, 92037, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| |
Collapse
|
942
|
Boeuf P, Drummer HE, Richards JS, Scoullar MJL, Beeson JG. The global threat of Zika virus to pregnancy: epidemiology, clinical perspectives, mechanisms, and impact. BMC Med 2016; 14:112. [PMID: 27487767 PMCID: PMC4973112 DOI: 10.1186/s12916-016-0660-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 07/27/2016] [Indexed: 12/26/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus that has newly emerged as a significant global threat, especially to pregnancy. Recent major outbreaks in the Pacific and in Central and South America have been associated with an increased incidence of microcephaly and other abnormalities of the central nervous system in neonates. The causal link between ZIKV infection during pregnancy and microcephaly is now strongly supported. Over 2 billion people live in regions conducive to ZIKV transmission, with ~4 million infections in the Americas predicted for 2016. Given the scale of the current pandemic and the serious and long-term consequences of infection during pregnancy, the impact of ZIKV on health services and affected communities could be enormous. This further highlights the need for a rapid global public health and research response to ZIKV to limit and prevent its impact through the development of therapeutics, vaccines, and improved diagnostics. Here we review the epidemiology of ZIKV; the threat to pregnancy; the clinical consequences and broader impact of ZIKV infections; and the virus biology underpinning new interventions, diagnostics, and insights into the mechanisms of disease.
Collapse
Affiliation(s)
- Phillipe Boeuf
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia.
- Department of Medicine, The University of Melbourne, Melbourne, Australia.
| | - Heidi E Drummer
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
- Department of Microbiology, Monash University, Clayton, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, Australia
| | - Jack S Richards
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
- Department of Microbiology, Monash University, Clayton, Australia
| | - Michelle J L Scoullar
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - James G Beeson
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia.
- Department of Medicine, The University of Melbourne, Melbourne, Australia.
- Department of Microbiology, Monash University, Clayton, Australia.
| |
Collapse
|
943
|
Klase ZA, Khakhina S, Schneider ADB, Callahan MV, Glasspool-Malone J, Malone R. Zika Fetal Neuropathogenesis: Etiology of a Viral Syndrome. PLoS Negl Trop Dis 2016; 10:e0004877. [PMID: 27560129 PMCID: PMC4999274 DOI: 10.1371/journal.pntd.0004877] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The ongoing Zika virus epidemic in the Americas and the observed association with both fetal abnormalities (primary microcephaly) and adult autoimmune pathology (Guillain-Barré syndrome) has brought attention to this neglected pathogen. While initial case studies generated significant interest in the Zika virus outbreak, larger prospective epidemiology and basic virology studies examining the mechanisms of Zika viral infection and associated pathophysiology are only now starting to be published. In this review, we analyze Zika fetal neuropathogenesis from a comparative pathology perspective, using the historic metaphor of "TORCH" viral pathogenesis to provide context. By drawing parallels to other viral infections of the fetus, we identify common themes and mechanisms that may illuminate the observed pathology. The existing data on the susceptibility of various cells to both Zika and other flavivirus infections are summarized. Finally, we highlight relevant aspects of the known molecular mechanisms of flavivirus replication.
Collapse
Affiliation(s)
- Zachary A Klase
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States of America
| | - Svetlana Khakhina
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania, United States of America
| | - Adriano De Bernardi Schneider
- Department of Bioinformatics and Genomics, University of North Carolina at Charlotte, Charlotte, North Carolina, United States of America
| | - Michael V Callahan
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Zika Foundation, College Station, Texas, United States of America
| | - Jill Glasspool-Malone
- Atheric Pharmaceutical, Scottsville, Virginia, United States of America
- Global Clinical Scholars Research Training Program, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Robert Malone
- Atheric Pharmaceutical, Scottsville, Virginia, United States of America
- Global Clinical Scholars Research Training Program, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
944
|
Hickman HD, Pierson TC. Zika in the Brain: New Models Shed Light on Viral Infection. Trends Mol Med 2016; 22:639-641. [PMID: 27345865 PMCID: PMC4990132 DOI: 10.1016/j.molmed.2016.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 06/08/2016] [Indexed: 11/22/2022]
Abstract
The current Zika virus (ZIKV) outbreak is associated with high numbers of human congenital birth defects, yet it has been unclear how ZIKV infection during pregnancy causes these abnormalities. Three new mouse models now show that ZIKV crosses the placenta and replicates in the brains of fetal mice.
Collapse
Affiliation(s)
- Heather D Hickman
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Theodore C Pierson
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
945
|
Abstract
[No Abstract Available].
Collapse
Affiliation(s)
- Hail M Al-Abdely
- General Directorate of Infection Prevention and Control, Ministry of Health, Kingdom of Saudi Arabia. E-mail.
| |
Collapse
|
946
|
Vadakkan KI. Neurodegenerative disorders share common features of "loss of function" states of a proposed mechanism of nervous system functions. Biomed Pharmacother 2016; 83:412-430. [PMID: 27424323 DOI: 10.1016/j.biopha.2016.06.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 06/07/2016] [Accepted: 06/25/2016] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are highly heterogeneous for the locations affected and the nature of the aggregated proteins. Nearly 80% of the neurodegenerative disorders occur sporadically, indicating that certain factors must combine to initiate the degenerative changes. The contiguous extension of degenerative changes from cell to cell, the association with viral fusion proteins, loss of dendritic spines (postsynaptic terminals), and the eventual degeneration of cells indicate the presence of a unique mechanism for inter-cellular spread of pathology. It is not known whether the "loss of function" states of the still unknown normal nervous system operations can lead to neurodegenerative disorders. Here, the possible loss of function states of a proposed normal nervous system function are examined. A reversible inter-postsynaptic functional LINK (IPL) mechanism, consisting of transient inter-postsynaptic membrane (IPM) hydration exclusion and partial to complete IPM hemifusions, was proposed as a critical step necessary for the binding process and the induction of internal sensations of higher brain functions. When various findings from different neurodegenerative disorders are systematically organized and examined, disease features match the effects of loss of function states of different IPLs. Changes in membrane composition, enlargement of dendritic spines by dopamine and viral fusion proteins are capable of altering the IPLs to form IPM fusion. The latter can lead to the observed lateral spread of pathology, inter-neuronal cytoplasmic content mixing and abnormal protein aggregation. Since both the normal mechanism of reversible IPM hydration exclusion and the pathological process of transient IPM fusion can evade detection, testing their occurrence may provide preventive and therapeutic opportunities for these disorders.
Collapse
|
947
|
Bona ACD, Chitolina RF, Fermino ML, de Castro Poncio L, Weiss A, Lima JBP, Paldi N, Bernardes ES, Henen J, Maori E. Larval application of sodium channel homologous dsRNA restores pyrethroid insecticide susceptibility in a resistant adult mosquito population. Parasit Vectors 2016; 9:397. [PMID: 27416771 PMCID: PMC4946210 DOI: 10.1186/s13071-016-1634-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/07/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mosquitoes host and pass on to humans a variety of disease-causing pathogens such as infectious viruses and other parasitic microorganisms. The emergence and spread of insecticide resistance is threatening the effectiveness of current control measures for common mosquito vector borne diseases, such as malaria, dengue and Zika. Therefore, the emerging resistance to the widely used pyrethroid insecticides is an alarming problem for public health. Herein we demonstrated the use of RNA interference (RNAi) to increase susceptibility of adult mosquitoes to a widely used pyrethroid insecticide. METHODS Experiments were performed on a field-collected pyrethroid resistant strain of Ae. aegypti (Rio de Janeiro; RJ). Larvae from the resistant Ae. aegypti population were soaked with double-stranded RNAs (dsRNAs) that correspond either to voltage-gate sodium channel (VGSC), P-glycoprotein, or P450 detoxification genes and reared to adulthood. Adult mortality rates in the presence of various Deltamethrin pyrethroid concentrations were used to assess mosquito insecticide susceptibility. RESULTS We characterized the RJ Ae. aegypti strain with regard to its level of resistance to a pyrethroid insecticide and found that it was approximately 6 times more resistant to Deltamethrin compared to the laboratory Rockefeller strain. The RJ strain displayed a higher frequency of Val1016Ile and Phe1534Cys substitutions of the VGSC gene. The resistant strain also displayed a higher basal expression level of VGSC compared to the Rockefeller strain. When dsRNA-treated mosquitoes were subjected to a standard pyrethroid contact bioassay, only dsRNA targeting VGSC increased the adult mortality of the pyrethroid resistant strain. The dsRNA treatment proved effective in increasing adult mosquito susceptibility over a range of pyrethroid concentrations and these results were associated with dsRNA-specific small interfering RNAs in treated adults, and the corresponding specific down regulation of VGSC gene expression level. Finally, we demonstrated that the efficiency of our approach was further improved by 'tiling' along the VGSC gene in order to identify the most potent dsRNA sequences. CONCLUSIONS These results demonstrate that dsRNA applied to mosquito larvae retains its biological activity into adulthood. Thus, the RNAi system reported here could be a useful approach to control the widespread insecticide resistance in mosquitoes and other insect vectors of human diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - José Bento Pereira Lima
- Instituto Oswaldo Cruz - Fiocruz, Laboratório de Fisiologia e Controle de artrópodes vetores, Rio de Janeiro, RJ, Brazil
| | | | - Emerson Soares Bernardes
- Forrest Brasil Tecnologia Ltda, Curitiba, PR, Brazil.,Nuclear Energy Research Institute, Radiopharmacy Center, São Paulo, Brazil
| | | | - Eyal Maori
- Forrest Innovations Ltd, Caesarea, Israel.
| |
Collapse
|
948
|
Tang BL. Zika virus as a causative agent for primary microencephaly: the evidence so far. Arch Microbiol 2016; 198:595-601. [PMID: 27412681 DOI: 10.1007/s00203-016-1268-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 06/30/2016] [Accepted: 07/08/2016] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) infection has been associated with congenital microcephaly and peripheral neuropathy. The ongoing epidemic has triggered swift responses in the scientific community, and a number of recent reports have now confirmed a causal relationship between ZIKV infection and birth defect. In particular, ZIKV has been shown to be capable of compromising and crossing the placental barrier and infect the developing fetal brain, resulting in the demise and functional impairment of neuroprogenitor cells critical for fetal cortex development. Here, the evidence for ZIKV as a teratogenic agent that causes microcephaly is reviewed, and its association with other disorders is discussed.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, 28 Medical Drive, Singapore, 117456, Singapore.
| |
Collapse
|
949
|
Kim K, Shresta S. Neuroteratogenic Viruses and Lessons for Zika Virus Models. Trends Microbiol 2016; 24:622-636. [PMID: 27387029 DOI: 10.1016/j.tim.2016.06.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/29/2016] [Accepted: 06/02/2016] [Indexed: 01/08/2023]
Abstract
The Centers for Disease Control and Prevention has confirmed that Zika virus (ZIKV) causes congenital microcephaly. ZIKV now joins five other neuroteratogenic (NT) viruses in humans and ZIKV research is in its infancy. In addition, there is only one other NT human arbovirus (Venezuelan equine encephalitis virus), which is also poorly understood. But further insight into ZIKV can be found by evaluating arboviruses in domestic animals, of which there are at least seven NT viruses, three of which have been well studied. Here we review two key anatomical structures involved in modeling transplacental NT virus transmission: the placenta and the fetal blood-brain barrier. We then survey major research findings regarding transmission of NT viruses for guidance in establishing a mouse model of Zika disease that is crucial for a better understanding of ZIKV transmission and pathogenesis.
Collapse
Affiliation(s)
- Kenneth Kim
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Sujan Shresta
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| |
Collapse
|
950
|
Martinez ME. Preventing Zika Virus Infection during Pregnancy Using a Seasonal Window of Opportunity for Conception. PLoS Biol 2016; 14:e1002520. [PMID: 27467271 PMCID: PMC4965063 DOI: 10.1371/journal.pbio.1002520] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
It has come to light that Zika virus (ZIKV) infection during pregnancy can result in trans-placental transmission to the fetus along with fetal death, congenital microcephaly, and/or Central Nervous System (CNS) malformations. There are projected to be >9,200,000 births annually in countries with ongoing ZIKV transmission. In response to the ZIKV threat, the World Health Organization (WHO) is strategically targeting prevention of infection in pregnant women and funding contraception in epidemic regions. I propose that the damaging effects of ZIKV can be reduced using a seasonal window of opportunity for conception that may minimize maternal exposure. Like other acute viral infections-including the related flavivirus, dengue virus (DENV)-the transmission of ZIKV is anticipated to be seasonal. By seasonally planning pregnancy, this aspect of pathogen ecology can be leveraged to align sensitive periods of gestation with the low-transmission season.
Collapse
Affiliation(s)
- Micaela Elvira Martinez
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, New Jersey, United States of America
- Global Health Program, Woodrow Wilson School of Public and International Affairs, Princeton University, Princeton, New Jersey, United States of America
| |
Collapse
|