51
|
Kuzmina NA, Younan P, Gilchuk P, Santos RI, Flyak AI, Ilinykh PA, Huang K, Lubaki NM, Ramanathan P, Crowe JE, Bukreyev A. Antibody-Dependent Enhancement of Ebola Virus Infection by Human Antibodies Isolated from Survivors. Cell Rep 2018; 24:1802-1815.e5. [PMID: 30110637 PMCID: PMC6697154 DOI: 10.1016/j.celrep.2018.07.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 06/12/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022] Open
Abstract
Some monoclonal antibodies (mAbs) recovered from survivors of filovirus infections can protect against infection. It is currently unknown whether natural infection also induces some antibodies with the capacity for antibody-dependent enhancement (ADE). A panel of mAbs obtained from human survivors of filovirus infection caused by Ebola, Bundibugyo, or Marburg viruses was evaluated for their ability to facilitate ADE. ADE was observed readily with all mAbs examined at sub-neutralizing concentrations, and this effect was not restricted to mAbs with a particular epitope specificity, neutralizing capacity, or subclass. Blocking of specific Fcγ receptors reduced but did not abolish ADE that was associated with high-affinity binding antibodies, suggesting that lower-affinity interactions still cause ADE. Mutations of Fc fragments of an mAb that altered its interaction with Fc receptors rendered the antibody partially protective in vivo at a low dose, suggesting that ADE counteracts antibody-mediated protection and facilitates dissemination of filovirus infections.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/isolation & purification
- Antibodies, Monoclonal/pharmacology
- Antibodies, Neutralizing/isolation & purification
- Antibodies, Neutralizing/pharmacology
- Antibodies, Viral/isolation & purification
- Antibodies, Viral/pharmacology
- Antibody-Dependent Enhancement
- Ebolavirus/drug effects
- Ebolavirus/genetics
- Ebolavirus/immunology
- Ebolavirus/pathogenicity
- Epitopes/genetics
- Epitopes/immunology
- Gene Expression
- Genes, Reporter
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Hemorrhagic Fever, Ebola/immunology
- Hemorrhagic Fever, Ebola/mortality
- Hemorrhagic Fever, Ebola/therapy
- Hemorrhagic Fever, Ebola/virology
- Humans
- Immune Sera/chemistry
- Immunoglobulin Fc Fragments/chemistry
- Immunoglobulin Fc Fragments/genetics
- Marburg Virus Disease/immunology
- Marburg Virus Disease/mortality
- Marburg Virus Disease/therapy
- Marburg Virus Disease/virology
- Marburgvirus/drug effects
- Marburgvirus/genetics
- Marburgvirus/pathogenicity
- Mice
- Mice, Inbred BALB C
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/virology
- Primary Cell Culture
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Survival Analysis
- Survivors
- THP-1 Cells
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Patrick Younan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rodrigo I Santos
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Andrew I Flyak
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA
| | - Philipp A Ilinykh
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Kai Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Ndongala M Lubaki
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Palaniappan Ramanathan
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
52
|
Carro AC, Piccini LE, Damonte EB. Blockade of dengue virus entry into myeloid cells by endocytic inhibitors in the presence or absence of antibodies. PLoS Negl Trop Dis 2018; 12:e0006685. [PMID: 30092029 PMCID: PMC6103515 DOI: 10.1371/journal.pntd.0006685] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 08/21/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022] Open
Abstract
Background Dengue is the most prevalent arthropod-borne viral human disease in tropical and subtropical regions, caused by four dengue virus (DENV) serotypes. In spite of the increasing global incidence, no specific antiviral therapy is available. Cells of the mononuclear phagocyte lineage are the main targets either for direct antibody (Ab)-independent or Ab-mediated human DENV infection, usually associated to the severe forms of disease. Since the virus entry may be a convenient therapeutic alternative, this study aimed to investigate the mode of DENV internalization into myeloid cells in the absence and presence of DENV Ab and evaluate the inhibitory activity of diverse biochemical inhibitors of endocytosis. Methodology/principal findings By infectivity assays and quantitative RT-PCR determinations, it was demonstrated that DENV-2 entry into U937 and K562 cells in the absence of Ab was highly inhibited by the early treatment with ammonium chloride, chlorpromazine and dynasore, but it was not affected by methyl-β-cyclodextrin, indicating that DENV-2 utilizes a low pH-dependent, clathrin- and dynamin-mediated endocytic infectious pathway for the direct entry into both human myeloid cells. To study the Ab-mediated entry of DENV, the experimental conditions for enhancement of infection were established by inoculating immune complexes formed with DENV-2 and the Ab 2H2 or 3H5. The internalization of DENV-2-2H2 or DENV-2-3H5 complexes in both myeloid cells was also dependent on acid pH and dynamin but a differential requirement of the clathrin-mediated endocytic route was observed depending on the FcγR involved in the complex uptake: the infection through FcγRII was dependent on clathrin-coated vesicles whereas the internalization pathway mediated by FcγRI was independent of clathrin. This property was not serotype-specific. Conclusions/significance DENV entry into myeloid cells in the absence or presence of Ab can be blocked by diverse biochemical inhibitors affecting the cellular factors involved in endocytosis. The identification of the virus-host interactions involved in virus penetration may allow the finding of host-targeted antivirals widely active against diverse pathogenic flaviviruses with similar requirements for virus entry. Dengue is currently a widespread viral disease transmitted to human by mosquitoes, with very high prevalence in tropical and subtropical regions of América and Asia. Approximately 2.5 billion people are living in endemic areas and it is estimated that 350 million apparent and inapparent infections occur each year. There is no specific antiviral for treatment of dengue patients. On this basis, the search of antiviral agents is an urgent need. Dengue virus (DENV) entry is an attractive alternative for chemotherapeutic intervention since it represents a barrier to block the beginning of infection. We intended to evaluate the antiviral activity of diverse biochemical inhibitors of endocytosis against DENV in human myeloid cells. Results showed that DENV entry into these cells can be blocked by diverse biochemical inhibitors affecting the cellular factors involved in endocytosis such as intravesicular pH, clathrin-coated vesicles and dynamin, although the presence of non neutralizing antibodies, as can occur in secondary human infections, may alter the entry pathway under certain conditions. Then, the blockade of virus entry with host-targeted inhibitors may be a promising strategy for the development of safe antiviral agents with wide spectrum of activity against DENV and other related pathogenic flaviviruses.
Collapse
Affiliation(s)
- Ana C Carro
- Laboratory of Virology, Department of Biological Chemistry, Faculty of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Luana E Piccini
- Laboratory of Virology, Department of Biological Chemistry, Faculty of Sciences, University of Buenos Aires, Buenos Aires, Argentina
- IQUIBICEN, National Research Council (CONICET)-Department of Biological Chemistry, University of Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Elsa B Damonte
- Laboratory of Virology, Department of Biological Chemistry, Faculty of Sciences, University of Buenos Aires, Buenos Aires, Argentina
- IQUIBICEN, National Research Council (CONICET)-Department of Biological Chemistry, University of Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| |
Collapse
|
53
|
Marzinek JK, Bag N, Huber RG, Holdbrook DA, Wohland T, Verma CS, Bond PJ. A Funneled Conformational Landscape Governs Flavivirus Fusion Peptide Interaction with Lipid Membranes. J Chem Theory Comput 2018; 14:3920-3932. [DOI: 10.1021/acs.jctc.8b00438] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jan K. Marzinek
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | | | - Roland G. Huber
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | - Daniel A. Holdbrook
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| | | | - Chandra S. Verma
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 63755
| | - Peter J. Bond
- Bioinformatics Institute (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671
| |
Collapse
|
54
|
Rodríguez Y, Rojas M, Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, Monsalve DM, Gershwin ME, Anaya JM. Guillain-Barré syndrome, transverse myelitis and infectious diseases. Cell Mol Immunol 2018; 15:547-562. [PMID: 29375121 PMCID: PMC6079071 DOI: 10.1038/cmi.2017.142] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Guillain-Barré syndrome (GBS) and transverse myelitis (TM) both represent immunologically mediated polyneuropathies of major clinical importance. Both are thought to have a genetic predisposition, but as of yet no specific genetic risk loci have been clearly defined. Both are considered autoimmune, but again the etiologies remain enigmatic. Both may be induced via molecular mimicry, particularly from infectious agents and vaccines, but clearly host factor and co-founding host responses will modulate disease susceptibility and natural history. GBS is an acute inflammatory immune-mediated polyradiculoneuropathy characterized by tingling, progressive weakness, autonomic dysfunction, and pain. Immune injury specifically takes place at the myelin sheath and related Schwann-cell components in acute inflammatory demyelinating polyneuropathy, whereas in acute motor axonal neuropathy membranes on the nerve axon (the axolemma) are the primary target for immune-related injury. Outbreaks of GBS have been reported, most frequently related to Campylobacter jejuni infection, however, other agents such as Zika Virus have been strongly associated. Patients with GBS related to infections frequently produce antibodies against human peripheral nerve gangliosides. In contrast, TM is an inflammatory disorder characterized by acute or subacute motor, sensory, and autonomic spinal cord dysfunction. There is interruption of ascending and descending neuroanatomical pathways on the transverse plane of the spinal cord similar to GBS. It has been suggested to be triggered by infectious agents and molecular mimicry. In this review, we will focus on the putative role of infectious agents as triggering factors of GBS and TM.
Collapse
Affiliation(s)
- Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Manuel Rojas
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yovana Pacheco
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California Davis, School of Medicine, Davis, USA, CA
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.
| |
Collapse
|
55
|
Morris G, Barichello T, Stubbs B, Köhler CA, Carvalho AF, Maes M. Zika Virus as an Emerging Neuropathogen: Mechanisms of Neurovirulence and Neuro-Immune Interactions. Mol Neurobiol 2018; 55:4160-4184. [PMID: 28601976 DOI: 10.1007/s12035-017-0635-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023]
Abstract
Zika virus (ZIKV) is an emerging arbovirus of the genus Flaviviridae, which causes a febrile illness and has spread from across the Pacific to the Americas in a short timeframe. Convincing evidence has implicated the ZIKV to incident cases of neonatal microcephaly and a set of neurodevelopmental abnormalities referred to as the congenital Zika virus syndrome. In addition, emerging data points to an association with the ZIKV and the development of the so-called Guillain-Barre syndrome, an acute autoimmune polyneuropathy. Accumulating knowledge suggests that neurovirulent strains of the ZIKV have evolved from less pathogenic lineages of the virus. Nevertheless, mechanisms of neurovirulence and host-pathogen neuro-immune interactions remain incompletely elucidated. This review provides a critical discussion of genetic and structural alterations in the ZIKV which could have contributed to the emergence of neurovirulent strains. In addition, a mechanistic framework of neuro-immune mechanisms related to the emergence of neuropathology after ZIKV infection is discussed. Recent advances in knowledge point to avenues for the development of a putative vaccine as well as novel therapeutic strategies. Nevertheless, there are unique unmet challenges that need to be addressed in this regard. Finally, a research agenda is proposed.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
| | - Tatiana Barichello
- Laboratory of Experimental Microbiology, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
- Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, Denmark Hill, London, SE5 8AZ, UK
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Bishop Hall Lane, Chelmsford, CM1 1SQ, UK
| | - Cristiano A Köhler
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, P.O. Box 291, Geelong, VIC, 3220, Australia.
- Health Sciences Postgraduate Program, Health Sciences Center, State University of Londrina, Londrina, Parana, Brazil.
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Revitalis, Waalre, The Netherlands.
- Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
56
|
Acosta-Ampudia Y, Monsalve DM, Castillo-Medina LF, Rodríguez Y, Pacheco Y, Halstead S, Willison HJ, Anaya JM, Ramírez-Santana C. Autoimmune Neurological Conditions Associated With Zika Virus Infection. Front Mol Neurosci 2018; 11:116. [PMID: 29695953 PMCID: PMC5904274 DOI: 10.3389/fnmol.2018.00116] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 03/26/2018] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus rapidly spreading throughout the tropical Americas. Aedes mosquitoes is the principal way of transmission of the virus to humans. ZIKV can be spread by transplacental, perinatal, and body fluids. ZIKV infection is often asymptomatic and those with symptoms present minor illness after 3 to 12 days of incubation, characterized by a mild and self-limiting disease with low-grade fever, conjunctivitis, widespread pruritic maculopapular rash, arthralgia and myalgia. ZIKV has been linked to a number of central and peripheral nervous system injuries such as Guillain-Barré syndrome (GBS), transverse myelitis (TM), meningoencephalitis, ophthalmological manifestations, and other neurological complications. Nevertheless, mechanisms of host-pathogen neuro-immune interactions remain incompletely elucidated. This review provides a critical discussion about the possible mechanisms underlying the development of autoimmune neurological conditions associated with Zika virus infection.
Collapse
Affiliation(s)
- Yeny Acosta-Ampudia
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Diana M Monsalve
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Luis F Castillo-Medina
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Yovana Pacheco
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Susan Halstead
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Hugh J Willison
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Carolina Ramírez-Santana
- Center for Autoimmune Diseases Research, School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| |
Collapse
|
57
|
Hernandez-Morales I, Geluykens P, Clynhens M, Strijbos R, Goethals O, Megens S, Verheyen N, Last S, McGowan D, Coesemans E, De Boeck B, Stoops B, Devogelaere B, Pauwels F, Vandyck K, Berke JM, Raboisson P, Simmen K, Lory P, Van Loock M. Characterization of a dengue NS4B inhibitor originating from an HCV small molecule library. Antiviral Res 2017; 147:149-158. [DOI: 10.1016/j.antiviral.2017.10.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 10/08/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022]
|
58
|
Wang TT, Sewatanon J, Memoli MJ, Wrammert J, Bournazos S, Bhaumik SK, Pinsky BA, Chokephaibulkit K, Onlamoon N, Pattanapanyasat K, Taubenberger JK, Ahmed R, Ravetch JV. IgG antibodies to dengue enhanced for FcγRIIIA binding determine disease severity. Science 2017; 355:395-398. [PMID: 28126818 DOI: 10.1126/science.aai8128] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/04/2017] [Indexed: 12/12/2022]
Abstract
Dengue virus (DENV) infection in the presence of reactive, non-neutralizing immunoglobulin G (IgG) (RNNIg) is the greatest risk factor for dengue hemorrhagic fever (DHF) or dengue shock syndrome (DSS). Progression to DHF/DSS is attributed to antibody-dependent enhancement (ADE); however, because only a fraction of infections occurring in the presence of RNNIg advance to DHF/DSS, the presence of RNNIg alone cannot account for disease severity. We discovered that DHF/DSS patients respond to infection by producing IgGs with enhanced affinity for the activating Fc receptor FcγRIIIA due to afucosylated Fc glycans and IgG1 subclass. RNNIg enriched for afucosylated IgG1 triggered platelet reduction in vivo and was a significant risk factor for thrombocytopenia. Thus, therapeutics and vaccines restricting production of afucosylated, IgG1 RNNIg during infection may prevent ADE of DENV disease.
Collapse
Affiliation(s)
- Taia T Wang
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.,Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jaturong Sewatanon
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.,Department of Microbiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand 10700
| | - Matthew J Memoli
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jens Wrammert
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.,Division of Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Siddhartha Kumar Bhaumik
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA.,Division of Infectious Disease, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Benjamin A Pinsky
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kulkanya Chokephaibulkit
- Department of Pediatrics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand 10700
| | - Nattawat Onlamoon
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand 10700
| | - Kovit Pattanapanyasat
- Department of Research and Development, Faculty of Medicine Siriraj Hospital, Bangkok, Thailand 10700
| | - Jeffery K Taubenberger
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rafi Ahmed
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA.,Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
59
|
Elaboration of tetravalent antibody responses against dengue viruses using a subunit vaccine comprised of a single consensus dengue envelope sequence. Vaccine 2017; 35:6308-6320. [PMID: 28987441 DOI: 10.1016/j.vaccine.2017.09.063] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/18/2017] [Accepted: 09/20/2017] [Indexed: 02/01/2023]
Abstract
Dengue viruses (DENVs) are re-emerging pathogens transmitted by mosquitoes mainly in tropical and subtropical regions. Each year, they are estimated to infect 390 million people globally. The major challenge confronting dengue vaccine development is the need to induce balanced, long lasting tetravalent immune responses against four co-circulating virus serotypes (DENV-I, -II, -III, -IV), because primary infection by any one of which may predispose infected individuals to more severe diseases during a heterotypic secondary infection. Another difficulty is to select representative strains in vaccine design to provide cross-protection against most circulating virus strains. In this study, aimed at developing a tetravalent subunit vaccine with a representative single protein, we designed two vaccines (named cE80(D4) and cE80(max)) based on the consensus sequences of the ectodomain of envelope protein of 3127 DENV strains, and then expressed them in the baculovirus expression system. Both vaccines were capable of eliciting specific antibodies against all four DENV serotypes, and the predominant IgG subtype elicited by the two vaccines was IgG1. Moreover, these vaccines activated both type I and type II antigen-specific helper T cells that secreted IFN-γ and IL-4, respectively. This proof-of-concept study has set foundation for further optimization of a single protein-based tetravalent DENV vaccine.
Collapse
|
60
|
Use of a Recombinant Gamma-2 Herpesvirus Vaccine Vector against Dengue Virus in Rhesus Monkeys. J Virol 2017; 91:JVI.00525-17. [PMID: 28592531 DOI: 10.1128/jvi.00525-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/28/2017] [Indexed: 12/31/2022] Open
Abstract
Research on vaccine approaches that can provide long-term protection against dengue virus infection is needed. Here we describe the construction, immunogenicity, and preliminary information on the protective capacity of recombinant, replication-competent rhesus monkey rhadinovirus (RRV), a persisting herpesvirus. One RRV construct expressed nonstructural protein 5 (NS5), while a second recombinant expressed a soluble variant of the E protein (E85) of dengue virus 2 (DENV2). Four rhesus macaques received a single vaccination with a mixture of both recombinant RRVs and were subsequently challenged 19 weeks later with 1 × 105 PFU of DENV2. During the vaccine phase, plasma of all vaccinated monkeys showed neutralizing activity against DENV2. Cellular immune responses against NS5 were also elicited, as evidenced by major histocompatibility complex class I (MHC-I) tetramer staining in the one vaccinated monkey that was Mamu-A*01 positive. Unlike two of two unvaccinated controls, two of the four vaccinated monkeys showed no detectable viral RNA sequences in plasma after challenge. One of these two monkeys also showed no anamnestic increases in antibody levels following challenge and thus appeared to be protected against the acquisition of DENV2 following high-dose challenge. Continued study will be needed to evaluate the performance of herpesviral and other persisting vectors for achieving long-term protection against dengue virus infection.IMPORTANCE Continuing studies of vaccine approaches against dengue virus (DENV) infection are warranted, particularly ones that may provide long-term immunity against all four serotypes. Here we investigated whether recombinant rhesus monkey rhadinovirus (RRV) could be used as a vaccine against DENV2 infection in rhesus monkeys. Upon vaccination, all animals generated antibodies capable of neutralizing DENV2. Two of four vaccinated monkeys showed no detectable viral RNA after subsequent high-dose DENV2 challenge at 19 weeks postvaccination. Furthermore, one of these vaccinated monkeys appeared to be protected against the acquisition of DENV2 infection on the basis of undetectable viral loads and the lack of an anamnestic antibody response. These findings underscore the potential utility of recombinant herpesviruses as vaccine vectors.
Collapse
|
61
|
Abstract
A key determinant for the survival of organisms is their capacity to recognize and respond efficiently to foreign antigens. This is largely accomplished by the orchestrated activity of the innate and adaptive branches of the immune system. Antibodies are specifically generated in response to foreign antigens, facilitating thereby the specific recognition of antigens of almost infinite diversity. Receptors specific for the Fc domain of antibodies, Fc receptors, are expressed on the surface of the various myeloid leukocyte populations and mediate the binding and recognition of antibodies by innate leukocytes. By directly linking the innate and the adaptive components of immunity, Fc receptors play a central role in host defense and the maintenance of tissue homeostasis through the induction of diverse proinflammatory, anti-inflammatory, and immunomodulatory processes that are initiated upon engagement by the Fc domain. In this chapter, we discuss the mechanisms that regulate Fc domain binding to the various types of Fc receptors and provide an overview of the astonishing diversity of effector functions that are mediated through Fc-FcR interactions on myeloid cells. Lastly, we discuss the impact of FcR-mediated interactions in the context of IgG-mediated inflammation, autoimmunity, susceptibility to infection, and responsiveness to antibody-based therapeutics.
Collapse
|
62
|
Londono-Renteria B, Marinez-Angarita JC, Troupin A, Colpitts TM. Role of Mast Cells in Dengue Virus Pathogenesis. DNA Cell Biol 2017; 36:423-427. [PMID: 28486041 DOI: 10.1089/dna.2017.3765] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Dengue is currently regarded as the most common arthropod-borne viral disease in tropical and subtropical areas, with an estimated 50-100 million infections occurring each year. Nearly all patients experience a self-limiting clinical course; however, the illness ranges from undifferentiated fever to severe hemorrhagic fever with or without shock syndrome complications. There are several immune cells associated with the pathogenesis of dengue virus (DENV) infection and systemic spread, including dendritic cells, macrophages, and mast cells (MCs). MCs are widely recognized for their immune functions and as cellular regulators of vascular integrity in human skin. Furthermore, these cells are able to detect DENV, which results in activation and degranulation of potent vasoactive mediators prestored in the granules. These mediators can act directly on vascular endothelium, increasing permeability and inducing vascular leakage. This review is designed to present an insight into the role of MCs during DENV infection and the dual functions in immune protection and contribution to the most severe forms of dengue.
Collapse
Affiliation(s)
| | | | - Andrea Troupin
- 3 Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, South Carolina
| | - Tonya M Colpitts
- 3 Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, South Carolina
| |
Collapse
|
63
|
Wang M, Yang F, Huang D, Huang Y, Zhang X, Wang C, Zhang S, Zhang R. Anti-Idiotypic Antibodies Specific to prM Monoantibody Prevent Antibody Dependent Enhancement of Dengue Virus Infection. Front Cell Infect Microbiol 2017; 7:157. [PMID: 28536674 PMCID: PMC5422453 DOI: 10.3389/fcimb.2017.00157] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/12/2017] [Indexed: 01/08/2023] Open
Abstract
Dengue virus (DENV) co-circulates as four serotypes (DENV1-4). Primary infection only leads to self-limited dengue fever. But secondary infection with another serotype carries a higher risk of increased disease severity, causing life-threatening dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS). Serotype cross-reactive antibodies facilitate DENV infection in Fc-receptor-bearing cells by promoting virus entry via Fcγ receptors (FcγR), a process known as antibody dependent enhancement (ADE). Most studies suggested that enhancing antibodies were mainly specific to the structural premembrane protein (prM) of DENV. However, there is still no effective drugs or vaccines to prevent ADE. In this study, we firstly confirmed that both DENV-2 infected human sera (anti-DENV-2) and DENV-2 prM monoclonal antibody (prM mAb) could significantly enhance DENV-1 infection in K562 cells. Then we developed anti-idiotypic antibodies (prM-AIDs) specific to prM mAb by immunizing of Balb/c mice. Results showed that these polyclonal antibodies can dramatically reduce ADE phenomenon of DENV-1 infection in K562 cells. To further confirm the anti-ADE effect of prM-AIDs in vivo, interferon-α and γ receptor-deficient mice (AG6) were used as the mouse model for DENV infection. We found that administration of DENV-2 prM mAb indeed caused a higher DENV-1 titer as well as interleukin-10 (IL-10) and alaninea minotransferase (ALT) in mice infected with DENV-1, similar to clinical ADE symptoms. But when we supplemented prM-AIDs to DENV-1 challenged AG6 mice, the viral titer, IL-10 and ALT were obviously decreased to the negative control level. Of note, the number of platelets in peripheral blood of prM-AIDs group were significantly increased at day 3 post infection with DENV-1 compared that of prM-mAb group. These results confirmed that our prM-AIDs could prevent ADE not only in vitro but also in vivo, suggested that anti-idiotypic antibodies might be a new choice to be considered to treat DENV infection.
Collapse
Affiliation(s)
- Miao Wang
- College of Life Science and Oceanography, Shenzhen UniversityShenzhen, China
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Fan Yang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Dana Huang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Yalan Huang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Xiaomin Zhang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Chao Wang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Shaohua Zhang
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| | - Renli Zhang
- College of Life Science and Oceanography, Shenzhen UniversityShenzhen, China
- Shenzhen Center for Disease Control and PreventionShenzhen, China
| |
Collapse
|
64
|
Holbrook MR. Historical Perspectives on Flavivirus Research. Viruses 2017; 9:E97. [PMID: 28468299 PMCID: PMC5454410 DOI: 10.3390/v9050097] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/13/2017] [Accepted: 04/21/2017] [Indexed: 12/21/2022] Open
Abstract
The flaviviruses are small single-stranded RNA viruses that are typically transmitted by mosquito or tick vectors. These "arboviruses" are found around the world and account for a significant number of cases of human disease. The flaviviruses cause diseases ranging from mild or sub-clinical infections to lethal hemorrhagic fever or encephalitis. In many cases, survivors of neurologic flavivirus infections suffer long-term debilitating sequelae. Much like the emergence of West Nile virus in the United States in 1999, the recent emergence of Zika virus in the Americas has significantly increased the awareness of mosquito-borne viruses. The diseases caused by several flaviviruses have been recognized for decades, if not centuries. However, there is still a lot that is unknown about the flaviviruses as the recent experience with Zika virus has taught us. The objective of this review is to provide a general overview and some historical perspective on several flaviviruses that cause significant human disease. In addition, available medical countermeasures and significant gaps in our understanding of flavivirus biology are also discussed.
Collapse
Affiliation(s)
- Michael R Holbrook
- NIAID Integrated Research Facility, 8200 Research Plaza, Ft. Detrick, Frederick, MD 21702, USA.
| |
Collapse
|
65
|
Liu P, Ridilla M, Patel P, Betts L, Gallichotte E, Shahidi L, Thompson NL, Jacobson K. Beyond attachment: Roles of DC-SIGN in dengue virus infection. Traffic 2017; 18:218-231. [PMID: 28128492 DOI: 10.1111/tra.12469] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 01/24/2017] [Accepted: 01/24/2017] [Indexed: 12/16/2022]
Abstract
Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), a C-type lectin expressed on the plasma membrane by human immature dendritic cells, is a receptor for numerous viruses including Ebola, SARS and dengue. A controversial question has been whether DC-SIGN functions as a complete receptor for both binding and internalization of dengue virus (DENV) or whether it is solely a cell surface attachment factor, requiring either hand-off to another receptor or a co-receptor for internalization. To examine this question, we used 4 cell types: human immature dendritic cells and NIH3T3 cells expressing either wild-type DC-SIGN or 2 internalization-deficient DC-SIGN mutants, in which either the 3 cytoplasmic internalization motifs are silenced by alanine substitutions or the cytoplasmic region is truncated. Using confocal and super-resolution imaging and high content single particle tracking, we investigated DENV binding, DC-SIGN surface transport, endocytosis, as well as cell infectivity. DC-SIGN was found colocalized with DENV inside cells suggesting hand-off at the plasma membrane to another receptor did not occur. Moreover, all 3 DC-SIGN molecules on NIH3T3 cells supported cell infection. These results imply the involvement of a co-receptor because cells expressing the internalization-deficient mutants could still be infected.
Collapse
Affiliation(s)
- Ping Liu
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Marc Ridilla
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Pratik Patel
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Laurie Betts
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Emily Gallichotte
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Lidea Shahidi
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nancy L Thompson
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ken Jacobson
- Department of Cell Biology and Physiology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
66
|
The Antigenic Structure of Zika Virus and Its Relation to Other Flaviviruses: Implications for Infection and Immunoprophylaxis. Microbiol Mol Biol Rev 2017; 81:81/1/e00055-16. [PMID: 28179396 DOI: 10.1128/mmbr.00055-16] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Zika virus was discovered ∼70 years ago in Uganda and maintained a low profile as a human disease agent in Africa and Asia. Only recently has it caused explosive outbreaks in previously unaffected regions, first in Oceania and then in the Americas since 2015. Of special concern is the newly identified link between congenital malformations (especially microcephaly) and Zika virus infections during pregnancy. At present, it is unclear whether Zika virus changed its pathogenicity or whether the huge number of infections allowed the recognition of a previously cryptic pathogenic property. The purpose of this review is to discuss recent data on the molecular antigenic structure of Zika virus in the context of antibody-mediated neutralization and antibody-dependent enhancement (ADE) of infection, a phenomenon that has been implicated in the development of severe disease caused by the related dengue viruses. Emphasis is given to epitopes of antibodies that potently neutralize Zika virus and also to epitopes that provide antigenic links to other important human-pathogenic flaviviruses such as dengue, yellow fever, West Nile, Japanese encephalitis, and tick-borne encephalitis viruses. The antigenic cross talk between Zika and dengue viruses appears to be of special importance, since they cocirculate in many regions of endemicity and sequential infections are likely to occur frequently. New insights into the molecular antigenic structure of Zika virus and flaviviruses in general have provided the foundation for great progress made in developing Zika virus vaccines and antibodies for passive immunization.
Collapse
|
67
|
|
68
|
Suwanmanee S, Luplertlop N. Immunopathogenesis of Dengue Virus-Induced Redundant Cell Death: Apoptosis and Pyroptosis. Viral Immunol 2016; 30:13-19. [PMID: 27860556 DOI: 10.1089/vim.2016.0092] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dengue virus infection is a self-limited condition, which is of particular importance in tropical and subtropical regions and for which no specific treatment or effective vaccine is available. There are several hypotheses explaining dengue pathogenesis. These usually refer to host immune responses, including antibody-dependent enhancement, cytokine expression, and dengue virus particles including NS1 protein, which lead to cell death by both apoptosis and pyroptosis. A clear understanding of the pathogenesis should facilitate the development of vaccines and therapies. This review focuses on the immunopathogenesis in relation to clinical manifestations and patterns of cell death, focusing on the pathogenesis of severe dengue.
Collapse
Affiliation(s)
- San Suwanmanee
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University , Bangkok, Thailand
| | - Natthanej Luplertlop
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University , Bangkok, Thailand
| |
Collapse
|
69
|
Tsai HP, Tsai YY, Lin IT, Kuo PH, Chang KC, Chen JC, Ko WC, Wang JR. Validation and Application of a Commercial Quantitative Real-Time Reverse Transcriptase-PCR Assay in Investigation of a Large Dengue Virus Outbreak in Southern Taiwan. PLoS Negl Trop Dis 2016; 10:e0005036. [PMID: 27732593 PMCID: PMC5061319 DOI: 10.1371/journal.pntd.0005036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/13/2016] [Indexed: 11/30/2022] Open
Abstract
Background Accurate, rapid, and early diagnosis of dengue virus (DENV) infections is essential for optimal clinical care. Here, we evaluated the efficacy of the quantitative real-time PCR (qRT-PCR)-LightMix dengue virus EC kit for DENV detection using samples from a dengue outbreak in Taiwan in 2015. Methods Sera from patients with suspected DENV infection were analyzed and compared using the LightMix kit, a Dengue NS1 Ag + Ab Combo kit for detection of NS1 antigen and DENV-specific IgM and IgG antibodies, and an “in-house” qualitative DENV-specific RT-PCR assay. Results A total of 8,989, 8,954, and 1581 samples were subjected to NS1 antigen detection, IgM and IgG detection, and LightMix assays, respectively. The LightMix assay yielded a linear curve for viral loads (VL) between 102 and 106 copies/reaction, and the minimum detection limits for DENV serotype 1 (DENV1) and DENV2, DENV3, and DENV4 were 1, 10, and 100 focus forming units (FFU)/mL, respectively. There was 88.9% concordance between the results obtained using the NS1 antigen combo kit and by LightMix analysis, and the diagnostic sensitivity and specificity of the two methods were 89.4 and 100%, and 84.7 and 100%, respectively. Notably, fatal cases were attributed to DENV2 infection, and 79.5% (27/34) of these cases occurred in patients ≥ 71 years of age. Among these older patients, 82.3% (14/17) were NS1/IgM/IgG (+/-/-), exhibiting VLs between 106–109 copies/mL, which was markedly higher than the rate observed in the other age groups. Conclusions The LightMix assay was effective for early diagnosis of DENV infection. Our data indicate that high VLs during primary infection in elderly patients may be a positive predictor for severe illness, and may contribute to high mortality rates. The LightMix dengue virus EC qRT-PCR assay is effective for early diagnosis of DENV infection. High viral loads during primary infection in elderly patients may comprise a positive predictor for severe illness, and may contribute to high mortality rates.
Collapse
Affiliation(s)
- Huey-Pin Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - You-Yuan Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - I-Ting Lin
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Pin-Hwa Kuo
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Kung-Chao Chang
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Jung-Chin Chen
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
- Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
- * E-mail:
| |
Collapse
|
70
|
Schmidt CW. Zika in the United States: How Are We Preparing? ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:A157-65. [PMID: 27581255 PMCID: PMC5010403 DOI: 10.1289/ehp.124-a157] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
71
|
Antibody-Dependent Enhancement of Dengue Virus Infection in Primary Human Macrophages; Balancing Higher Fusion against Antiviral Responses. Sci Rep 2016; 6:29201. [PMID: 27380892 PMCID: PMC4933910 DOI: 10.1038/srep29201] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 06/14/2016] [Indexed: 12/13/2022] Open
Abstract
The dogma is that the human immune system protects us against pathogens. Yet, several viruses, like dengue virus, antagonize the hosts’ antibodies to enhance their viral load and disease severity; a phenomenon called antibody-dependent enhancement of infection. This study offers novel insights in the molecular mechanism of antibody-mediated enhancement (ADE) of dengue virus infection in primary human macrophages. No differences were observed in the number of bound and internalized DENV particles following infection in the absence and presence of enhancing concentrations of antibodies. Yet, we did find an increase in membrane fusion activity during ADE of DENV infection. The higher fusion activity is coupled to a low antiviral response early in infection and subsequently a higher infection efficiency. Apparently, subtle enhancements early in the viral life cycle cascades into strong effects on infection, virus production and immune response. Importantly, and in contrast to other studies, the antibody-opsonized virus particles do not trigger immune suppression and remain sensitive to interferon. Additionally, this study gives insight in how human macrophages interact and respond to viral infections and the tight regulation thereof under various conditions of infection.
Collapse
|
72
|
Santos Souza HF, da Silva Almeida B, Boscardin SB. Early dengue virus interactions: the role of dendritic cells during infection. Virus Res 2016; 223:88-98. [PMID: 27381061 DOI: 10.1016/j.virusres.2016.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 10/21/2022]
Abstract
Dengue is an acute infectious disease caused by dengue virus (DENV) that affects approximately 400 million people annually, being the most prevalent human arthropod-borne disease. DENV infection causes a wide variety of clinical manifestations that range from asymptomatic to dengue fever, and in some cases may evolve to the more severe dengue hemorrhagic fever and dengue shock syndrome. The exact reasons why some patients do not have symptoms while others develop the severe forms of disease are still elusive, but gathered evidence showed correlation between a secondary infection with a heterologous DENV serotype and the occurrence of severe symptoms. Despite several advances, the mechanisms of DENV infection are still not completely elucidated, and efforts have been made to understand the development of immunity and/or pathology to DENV. When a mosquito transmits DENV, the virus is initially deposited in the skin, where mononuclear phagocytic cells, such as dendritic cells (DCs), become infected. DCs play a critical role in the induction of immune responses, as they are able to rapidly detect pathogen-associated molecular patterns, endocytose and process antigens, and efficiently activate naïve-T and B cells. Recent findings have shown that DCs serve as DENV targets, but they are also important mediators of immunity against the virus. In this review, we will briefly discuss DENV infection pathogenesis, and introduce DCs as central players in the induction of anti-DENV immune responses. Then, we will review in more detail how DENV interacts with and is sensed by DCs, with particular emphasis in two classes of receptors implicated in viral entry.
Collapse
Affiliation(s)
- Higo Fernando Santos Souza
- Laboratory of Antigen Targeting Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bianca da Silva Almeida
- Laboratory of Antigen Targeting Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Silvia Beatriz Boscardin
- Laboratory of Antigen Targeting Dendritic Cells, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; National Institute of Science and Technology in Vaccines, Belo Horizonte, Brazil.
| |
Collapse
|
73
|
Lee PX, Ong LC, Libau EA, Alonso S. Relative Contribution of Dengue IgG Antibodies Acquired during Gestation or Breastfeeding in Mediating Dengue Disease Enhancement and Protection in Type I Interferon Receptor-Deficient Mice. PLoS Negl Trop Dis 2016; 10:e0004805. [PMID: 27341339 PMCID: PMC4920417 DOI: 10.1371/journal.pntd.0004805] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/04/2016] [Indexed: 01/22/2023] Open
Abstract
Dengue virus (DENV) causes a spectrum of diseases ranging from self-limiting dengue fever to severe conditions such as haemorrhagic fever and dengue shock syndrome. Antibody-dependent enhancement (ADE) is thought to explain the occurrence of severe dengue whereby pre-existing binding but non-neutralising antibodies enhance DENV infection. The ADE phenomenon is supported by epidemiological findings that infants that born to dengue immune mothers are at greater risk to develop severe dengue upon primary infection. The role of maternally acquired dengue-specific antibodies in disease enhancement was recently recapitulated in a mouse model where mice born to DENV1-immune mothers experienced enhanced disease severity upon DENV2 infection. Here, this study investigates the relative contribution of maternal dengue-specific antibodies acquired during gestation and breastfeeding in dengue disease. Using a surrogate breastfeeding mother experimental approach, we showed that majority of the maternal dengue-specific antibodies were acquired during breastfeeding and conferred an extended enhancement window. On the other hand, in the context of homologous infection, breastfeeding conferred protection. Furthermore, measurement of dengue-specific antibody titres over time in mice born to dengue immune mothers revealed a biphasic pattern of antibody decay as reported in humans. Our work provides evidence of the potential contribution of breast milk-acquired dengue-specific IgG antibodies in enhancement and protection against dengue. Should such contribution be established in humans as well, it may have important implications for the development of guidelines to dengue-immune breastfeeding mothers. Epidemiological observations showed that 5–9 month old infants born to dengue immune mothers have increased risk of developing severe disease upon primary dengue infection. This disease enhancement has been associated with the presence of binding but non-neutralizing maternal dengue antibodies. The recent development of experimental dengue mouse models involving maternal antibodies supports their role in both disease enhancement and protection. Here, we examined the contribution of maternal antibodies acquired during gestation and breastfeeding in disease enhancement and protection. Our findings support that majority of maternal IgG antibodies circulating in mice born to dengue immune mothers are acquired from breast milk. As such, we showed that breastfeeding conferred extended window of enhancement or protection. These findings provide the first experimental evidence for a role of breast milk dengue antibodies in mediating dengue infection outcome. This may help develop guidelines to dengue immune breastfeeding mothers.
Collapse
Affiliation(s)
- Pei Xuan Lee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Li Ching Ong
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Eshele Anak Libau
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Sylvie Alonso
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
74
|
Troupin A, Londono-Renteria B, Conway MJ, Cloherty E, Jameson S, Higgs S, Vanlandingham DL, Fikrig E, Colpitts TM. A novel mosquito ubiquitin targets viral envelope protein for degradation and reduces virion production during dengue virus infection. Biochim Biophys Acta Gen Subj 2016; 1860:1898-909. [PMID: 27241849 PMCID: PMC4949077 DOI: 10.1016/j.bbagen.2016.05.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 04/27/2016] [Accepted: 05/26/2016] [Indexed: 11/17/2022]
Abstract
Background Dengue virus (DENV) is a mosquito-borne flavivirus that causes significant human disease and mortality in the tropics and subtropics. By examining the effects of virus infection on gene expression, and interactions between virus and vector, new targets for prevention of infection and novel treatments may be identified in mosquitoes. We previously performed a microarray analysis of the Aedes aegypti transcriptome during infection with DENV and found that mosquito ubiquitin protein Ub3881 (AAEL003881) was specifically and highly down-regulated. Ubiquitin proteins have multiple functions in insects, including marking proteins for proteasomal degradation, regulating apoptosis and mediating innate immune signaling. Methods We used qRT-PCR to quantify gene expression and infection, and RNAi to reduce Ub3881 expression. Mosquitoes were infected with DENV through blood feeding. We transfected DENV protein expression constructs to examine the effect of Ub3881 on protein degradation. We used site-directed mutagenesis and transfection to determine what amino acids are involved in Ub3881-mediated protein degradation. Immunofluorescence, Co-immunoprecipitation and Western blotting were used to examine protein interactions and co-localization. Results The overexpression of Ub3881, but not related ubiquitin proteins, decreased DENV infection in mosquito cells and live Ae. aegypti. The Ub3881 protein was demonstrated to be involved in DENV envelope protein degradation and reduce the number of infectious virions released. Conclusions We conclude that Ub3881 has several antiviral functions in the mosquito, including specific viral protein degradation. General significance Our data highlights Ub3881 as a target for future DENV prevention strategies in the mosquito transmission vector. A novel mosquito ubiquitin, Ub3881, is identified in Aedes aegypti. Ub3881 is shown to have antiviral functions during dengue virus infection of the mosquito. Ub3881 targets a dengue viral protein for degradation during infection. Future dengue virus prevention strategies could incorporate Ub3881 as a target to prevent mosquito infection.
Collapse
Affiliation(s)
- Andrea Troupin
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Berlin Londono-Renteria
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States
| | - Michael J Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI 48859, United States
| | - Erin Cloherty
- Department of Tropical Medicine, Tulane University School of Public Health, New Orleans, LA 70112, United States
| | - Samuel Jameson
- Department of Tropical Medicine, Tulane University School of Public Health, New Orleans, LA 70112, United States
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, KS 66506, United States; Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Dana L Vanlandingham
- Biosecurity Research Institute, Kansas State University, Manhattan, KS 66506, United States; Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS 66506, United States
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, United States; Howard Hughes Medical Institute, Chevy Chase, MD 20815, United States
| | - Tonya M Colpitts
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, United States.
| |
Collapse
|
75
|
Vitamin D-Regulated MicroRNAs: Are They Protective Factors against Dengue Virus Infection? Adv Virol 2016; 2016:1016840. [PMID: 27293435 PMCID: PMC4879221 DOI: 10.1155/2016/1016840] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/07/2016] [Accepted: 04/20/2016] [Indexed: 12/19/2022] Open
Abstract
Over the last few years, an increasing body of evidence has highlighted the critical participation of vitamin D in the regulation of proinflammatory responses and protection against many infectious pathogens, including viruses. The activity of vitamin D is associated with microRNAs, which are fine tuners of immune activation pathways and provide novel mechanisms to avoid the damage that arises from excessive inflammatory responses. Severe symptoms of an ongoing dengue virus infection and disease are strongly related to highly altered production of proinflammatory mediators, suggesting impairment in homeostatic mechanisms that control the host's immune response. Here, we discuss the possible implications of emerging studies anticipating the biological effects of vitamin D and microRNAs during the inflammatory response, and we attempt to extrapolate these findings to dengue virus infection and to their potential use for disease management strategies.
Collapse
|
76
|
Martínez Gómez JM, Ong LC, Lam JH, Binte Aman SA, Libau EA, Lee PX, St. John AL, Alonso S. Maternal Antibody-Mediated Disease Enhancement in Type I Interferon-Deficient Mice Leads to Lethal Disease Associated with Liver Damage. PLoS Negl Trop Dis 2016; 10:e0004536. [PMID: 27007501 PMCID: PMC4805191 DOI: 10.1371/journal.pntd.0004536] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/22/2016] [Indexed: 11/18/2022] Open
Abstract
Epidemiological studies have reported that most of the severe dengue cases occur upon a secondary heterologous infection. Furthermore, babies born to dengue immune mothers are at greater risk of developing severe disease upon primary infection with a heterologous or homologous dengue virus (DENV) serotype when maternal antibodies reach sub-neutralizing concentrations. These observations have been explained by the antibody mediated disease enhancement (ADE) phenomenon whereby heterologous antibodies or sub-neutralizing homologous antibodies bind to but fail to neutralize DENV particles, allowing Fc-receptor mediated entry of the virus-antibody complexes into host cells. This eventually results in enhanced viral replication and heightened inflammatory responses. In an attempt to replicate this ADE phenomenon in a mouse model, we previously reported that upon DENV2 infection 5-week old type I and II interferon (IFN) receptors-deficient mice (AG129) born to DENV1-immune mothers displayed enhancement of disease severity characterized by increased virus titers and extensive vascular leakage which eventually led to the animals' death. However, as dengue occurs in immune competent individuals, we sought to reproduce this mouse model in a less immunocompromised background. Here, we report an ADE model that is mediated by maternal antibodies in type I IFN receptor-deficient A129 mice. We show that 5-week old A129 mice born to DENV1-immune mothers succumbed to a DENV2 infection within 4 days that was sub-lethal in mice born to naïve mothers. Clinical manifestations included extensive hepatocyte vacuolation, moderate vascular leakage, lymphopenia, and thrombocytopenia. Anti-TNFα therapy totally protected the mice and correlated with healthy hepatocytes. In contrast, blocking IL-6 did not impact the virus titers or disease outcome. This A129 mouse model of ADE may help dissecting the mechanisms involved in dengue pathogenesis and evaluate the efficacy of vaccine and therapeutic candidates.
Collapse
Affiliation(s)
- Julia María Martínez Gómez
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Li Ching Ong
- Infectious Disease programme, Singapore-MIT alliance for Research and Technology (SMART), National University of Singapore, Singapore
| | - Jian Hang Lam
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Eshele Anak Libau
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | - Pei Xuan Lee
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Sylvie Alonso
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Immunology programme, Life Sciences Institute, National University of Singapore, Singapore
- Infectious Disease programme, Singapore-MIT alliance for Research and Technology (SMART), National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
77
|
Yang H, Li Z, Lin H, Wang W, Yang J, Liu L, Zeng X, Wu Y, Yu Y, Li Y. A novel dengue virus serotype 1 vaccine candidate based on Japanese encephalitis virus vaccine strain SA14-14-2 as the backbone. Arch Virol 2016; 161:1517-26. [PMID: 26976137 DOI: 10.1007/s00705-016-2817-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/03/2016] [Indexed: 01/08/2023]
|
78
|
Huang X, Yue Y, Li D, Zhao Y, Qiu L, Chen J, Pan Y, Xi J, Wang X, Sun Q, Li Q. Antibody-dependent enhancement of dengue virus infection inhibits RLR-mediated Type-I IFN-independent signalling through upregulation of cellular autophagy. Sci Rep 2016; 6:22303. [PMID: 26923481 PMCID: PMC4770412 DOI: 10.1038/srep22303] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/11/2016] [Indexed: 12/25/2022] Open
Abstract
Antibody dependent enhancement (ADE) of dengue virus (DENV) infection is identified as the main risk factor of severe Dengue diseases. Through opsonization by subneutralizing or non-neutralizing antibodies, DENV infection suppresses innate cell immunity to facilitate viral replication. However, it is largely unknown whether suppression of type-I IFN is necessary for a successful ADE infection. Here, we report that both DENV and DENV-ADE infection induce an early ISG (NOS2) expression through RLR-MAVS signalling axis independent of the IFNs signaling. Besides, DENV-ADE suppress this early antiviral response through increased autophagy formation rather than induction of IL-10 secretion. The early induced autophagic proteins ATG5-ATG12 participate in suppression of MAVS mediated ISGs induction. Our findings suggest a mechanism for DENV to evade the early antiviral response before IFN signalling activation. Altogether, these results add knowledge about the complexity of ADE infection and contribute further to research on therapeutic strategies.
Collapse
Affiliation(s)
- Xinwei Huang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
- Key Laboratory of The Second Affiliated Hospital of Kuming Medical College, Kunming 650101, PR China
| | - Yaofei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Duo Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yujiao Zhao
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Lijuan Qiu
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Junying Chen
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Yue Pan
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Juemin Xi
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Xiaodan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| | - Qihan Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences, and Peking Union Medical College, Kunming 650118, PR China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming 650118, PR China
| |
Collapse
|
79
|
Talarico LB, Damonte EB. Characterization of in vitro dengue virus resistance to carrageenan. J Med Virol 2016; 88:1120-9. [DOI: 10.1002/jmv.24457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2015] [Indexed: 11/07/2022]
Affiliation(s)
- Laura B. Talarico
- Laboratory of Virology; Faculty of Sciences; Department of Biological Chemistry; University of Buenos Aires; Buenos Aires Argentina
| | - Elsa B. Damonte
- Laboratory of Virology; Faculty of Sciences; Department of Biological Chemistry; University of Buenos Aires; Buenos Aires Argentina
- IQUIBICEN-National Research Council (CONICET); Ciudad Universitaria; Buenos Aires Argentina
| |
Collapse
|
80
|
Tittarelli E, Barrero PR, Mistchenko AS, Valinotto LE. Secondary dengue virus infections during the 2009 outbreak in Buenos Aires. Trop Med Int Health 2015; 21:28-32. [PMID: 26458156 DOI: 10.1111/tmi.12619] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVES To evaluate the occurrence of secondary dengue virus (DENV) infections during the 2009 outbreak in a non-endemic area. Viral loads were evaluated in serum from acute-phase patients, comparing primary and secondary infection. METHODS Serum samples from patients with clinical diagnosis of suspected dengue were referred to the Virology Laboratory at 'Ricardo Gutiérrez' Children's Hospital. Dengue-positive samples were classified as primary or secondary DENV infections through serological methods (anti-DENV IgM and IgG). Viral loads were measured by quantitative real-time PCR (qRT-PCR) in samples obtained in the first 5 days of infection. Statistical analyses were performed to evaluate factors that might correlate with differences in the viral load of primary or secondary infection. RESULTS A total of 229 DENV cases were confirmed; among them, 22.7% were secondary infections. No significant differences were found between the viral load of primary and secondary infections. CONCLUSION We detected a high percentage of secondary DENV infections in a non-endemic area; this finding might correspond to socio-demographic characteristics of the group under study or indicate a previous cryptic DENV circulation causing inapparent infections.
Collapse
Affiliation(s)
- E Tittarelli
- Laboratorio de Virología Hospital de Niños 'Ricardo Gutiérrez', Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - P R Barrero
- Laboratorio de Virología Hospital de Niños 'Ricardo Gutiérrez', Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - A S Mistchenko
- Laboratorio de Virología Hospital de Niños 'Ricardo Gutiérrez', Buenos Aires, Argentina.,Comisión de Investigaciones Científicas de la Provincia de Buenos Aires, Buenos Aires, Argentina
| | - L E Valinotto
- Laboratorio de Virología Hospital de Niños 'Ricardo Gutiérrez', Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
81
|
Londono-Renteria B, Troupin A, Conway MJ, Vesely D, Ledizet M, Roundy CM, Cloherty E, Jameson S, Vanlandingham D, Higgs S, Fikrig E, Colpitts TM. Dengue Virus Infection of Aedes aegypti Requires a Putative Cysteine Rich Venom Protein. PLoS Pathog 2015; 11:e1005202. [PMID: 26491875 PMCID: PMC4619585 DOI: 10.1371/journal.ppat.1005202] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-borne flavivirus that causes serious human disease and mortality worldwide. There is no specific antiviral therapy or vaccine for DENV infection. Alterations in gene expression during DENV infection of the mosquito and the impact of these changes on virus infection are important events to investigate in hopes of creating new treatments and vaccines. We previously identified 203 genes that were ≥5-fold differentially upregulated during flavivirus infection of the mosquito. Here, we examined the impact of silencing 100 of the most highly upregulated gene targets on DENV infection in its mosquito vector. We identified 20 genes that reduced DENV infection by at least 60% when silenced. We focused on one gene, a putative cysteine rich venom protein (SeqID AAEL000379; CRVP379), whose silencing significantly reduced DENV infection in Aedes aegypti cells. Here, we examine the requirement for CRVP379 during DENV infection of the mosquito and investigate the mechanisms surrounding this phenomenon. We also show that blocking CRVP379 protein with either RNAi or specific antisera inhibits DENV infection in Aedes aegypti. This work identifies a novel mosquito gene target for controlling DENV infection in mosquitoes that may also be used to develop broad preventative and therapeutic measures for multiple flaviviruses.
Collapse
Affiliation(s)
- Berlin Londono-Renteria
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Andrea Troupin
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
| | - Michael J Conway
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, Michigan, United States of America
| | - Diana Vesely
- L2 Diagnostics, New Haven, Connecticut, United States of America
| | - Michael Ledizet
- L2 Diagnostics, New Haven, Connecticut, United States of America
| | - Christopher M. Roundy
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Erin Cloherty
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Samuel Jameson
- Department of Tropical Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, United States of America
| | - Dana Vanlandingham
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Stephen Higgs
- Biosecurity Research Institute, Kansas State University, Manhattan, Kansas, United States of America
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Tonya M. Colpitts
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States of America
- * E-mail:
| |
Collapse
|
82
|
Poggianella M, Slon Campos JL, Chan KR, Tan HC, Bestagno M, Ooi EE, Burrone OR. Dengue E Protein Domain III-Based DNA Immunisation Induces Strong Antibody Responses to All Four Viral Serotypes. PLoS Negl Trop Dis 2015. [PMID: 26218926 PMCID: PMC4517776 DOI: 10.1371/journal.pntd.0003947] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dengue virus (DENV) infection is a major emerging disease widely distributed throughout the tropical and subtropical regions of the world affecting several millions of people. Despite constants efforts, no specific treatment or effective vaccine is yet available. Here we show a novel design of a DNA immunisation strategy that resulted in the induction of strong antibody responses with high neutralisation titres in mice against all four viral serotypes. The immunogenic molecule is an engineered version of the domain III (DIII) of the virus E protein fused to the dimerising CH3 domain of the IgG immunoglobulin H chain. The DIII sequences were also codon-optimised for expression in mammalian cells. While DIII alone is very poorly secreted, the codon-optimised fusion protein is rightly expressed, folded and secreted at high levels, thus inducing strong antibody responses. Mice were immunised using gene-gun technology, an efficient way of intradermal delivery of the plasmid DNA, and the vaccine was able to induce neutralising titres against all serotypes. Additionally, all sera showed reactivity to a recombinant DIII version and the recombinant E protein produced and secreted from mammalian cells in a mono-biotinylated form when tested in a conformational ELISA. Sera were also highly reactive to infective viral particles in a virus-capture ELISA and specific for each serotype as revealed by the low cross-reactive and cross-neutralising activities. The serotype specific sera did not induce antibody dependent enhancement of infection (ADE) in non-homologous virus serotypes. A tetravalent immunisation protocol in mice showed induction of neutralising antibodies against all four dengue serotypes as well. Dengue disease is a mosquito-borne viral infection caused by Dengue virus (DENV), one of the most important human pathogens worldwide. DENV infection produces a systemic disease with a broad symptomatic spectrum ranging from mild febrile illness (Dengue Fever, DF) to severe haemorrhagic manifestations (Dengue Haemorrhagic fever and Dengue Shock Syndrome, DHF and DSS respectively). To date there is no vaccine available to prevent dengue disease. We show here a strategy of immunisation, tested in mice, that elicits a strong immune response against the four different DENV serotypes. The novelties presented in our work open the way to the development of an efficient vaccine accessible to developing countries.
Collapse
Affiliation(s)
- Monica Poggianella
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - José L. Slon Campos
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Kuan Rong Chan
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Hwee Cheng Tan
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Marco Bestagno
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Oscar R. Burrone
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
- * E-mail:
| |
Collapse
|
83
|
Nizard M, Diniz MO, Roussel H, Tran T, Ferreira LC, Badoual C, Tartour E. Mucosal vaccines: novel strategies and applications for the control of pathogens and tumors at mucosal sites. Hum Vaccin Immunother 2015; 10:2175-87. [PMID: 25424921 DOI: 10.4161/hv.29269] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.
Collapse
Affiliation(s)
- Mevyn Nizard
- a INSERM U970; Universite Paris Descartes; Sorbonne Paris-Cité; Paris, France
| | | | | | | | | | | | | |
Collapse
|
84
|
Abstract
Dengue is the most prevalent mosquito-borne viral disease worldwide. Yet, there are no vaccines or specific antivirals available to prevent or treat the disease. Several dengue vaccines are currently in clinical or preclinical stages. The most advanced vaccine is the chimeric tetravalent CYD-TDV vaccine of Sanofi Pasteur. This vaccine has recently cleared Phase III, and efficacy results have been published. Excellent tetravalent seroconversion was seen, yet the protective efficacy against infection was surprisingly low. Here, we will describe the complicating factors involved in the generation of a safe and efficacious dengue vaccine. Furthermore, we will discuss the human antibody responses during infection, including the epitopes targeted in humans. Also, we will discuss the current understanding of the assays used to evaluate antibody response. We hope this review will aid future dengue vaccine development as well as fundamental research related to the phenomenon of antibody-dependent enhancement of dengue virus infection.
Collapse
Affiliation(s)
- Jacky Flipse
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jolanda M. Smit
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
85
|
Castilla V, Piccini LE, Damonte EB. Dengue virus entry and trafficking: perspectives as antiviral target for prevention and therapy. Future Virol 2015. [DOI: 10.2217/fvl.15.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ABSTRACT Dengue virus (DENV) is the etiological agent of the most important human viral infection transmitted by mosquitoes in the world. In spite of the serious health threat that dengue represents, at present there are no vaccine or antiviral agents available and treatment of patients consists of supportive therapy. This review will focus on the process of DENV entry into the host cell as a potential target for antiviral therapy. The recent advances in the knowledge of viral and cellular molecules and mechanisms involved in binding, internalization and trafficking of DENV into the host cell until virion uncoating are discussed, together with an overview of the strategies and compounds evaluated for development of antiviral agents targeted to DENV entry.
Collapse
Affiliation(s)
- Viviana Castilla
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, Piso 4, 1428 Buenos Aires, Argentina
| | - Luana E Piccini
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, Piso 4, 1428 Buenos Aires, Argentina
| | - Elsa B Damonte
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pabellón 2, Piso 4, 1428 Buenos Aires, Argentina
| |
Collapse
|
86
|
Mathew A, Townsley E, Ennis FA. Elucidating the role of T cells in protection against and pathogenesis of dengue virus infections. Future Microbiol 2015; 9:411-25. [PMID: 24762312 DOI: 10.2217/fmb.13.171] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Dengue viruses (DENV) cause significantly more human disease than any other arbovirus, with hundreds of thousands of cases leading to severe disease in thousands annually. Antibodies and T cells induced by primary infection with DENV have the potential for both positive (protective) and negative (pathological) effects during subsequent DENV infections. In this review, we summarize studies that have examined T-cell responses in humans following natural infection and vaccination. We discuss studies that support a role for T cells in protection against and those that support a role for the involvement of T cells in the pathogenesis of severe disease. The mechanisms that lead to severe disease are complex, and T-cell responses are an important component that needs to be further evaluated for the development of safe and efficacious DENV vaccines.
Collapse
Affiliation(s)
- Anuja Mathew
- Division of Infectious Diseases & Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | |
Collapse
|
87
|
Novel recombinant chimeric virus-like particle is immunogenic and protective against both enterovirus 71 and coxsackievirus A16 in mice. Sci Rep 2015; 5:7878. [PMID: 25597595 PMCID: PMC4297979 DOI: 10.1038/srep07878] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/15/2014] [Indexed: 12/02/2022] Open
Abstract
Hand-foot-and-mouth disease (HFMD) has been recognized as an important global public health issue, which is predominantly caused by enterovirus 71 (EV-A71) and coxsackievirus A16 (CVA16). There is no available vaccine against HFMD. An ideal HFMD vaccine should be bivalent against both EV-A71 and CVA16. Here, a novel strategy to produce bivalent HFMD vaccine based on chimeric EV-A71 virus-like particles (ChiEV-A71 VLPs) was proposed and illustrated. The neutralizing epitope SP70 within the capsid protein VP1 of EV-A71 was replaced with that of CVA16 in ChiEV-A71 VLPs. Structural modeling revealed that the replaced CVA16-SP70 epitope is well exposed on the surface of ChiEV-A71 VLPs. These VLPs produced in Saccharomyces cerevisiae exhibited similarity in both protein composition and morphology as naive EV-A71 VLPs. Immunization with ChiEV-A71 VLPs in mice elicited robust Th1/Th2 dependent immune responses against EV-A71 and CVA16. Furthermore, passive immunization with anti-ChiEV-A71 VLPs sera conferred full protection against lethal challenge of both EV-A71 and CVA16 infection in neonatal mice. These results suggested that this chimeric vaccine, ChiEV-A71 might have the potential to be further developed as a bivalent HFMD vaccine in the near future. Such chimeric enterovirus VLPs provide an alternative platform for bivalent HFMD vaccine development.
Collapse
|
88
|
Parreira R, Sousa CA. Dengue fever in Europe: could there be an epidemic in the future? Expert Rev Anti Infect Ther 2014; 13:29-40. [DOI: 10.1586/14787210.2015.982094] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
89
|
Corbett KS, Katzelnick L, Tissera H, Amerasinghe A, de Silva AD, de Silva AM. Preexisting neutralizing antibody responses distinguish clinically inapparent and apparent dengue virus infections in a Sri Lankan pediatric cohort. J Infect Dis 2014; 211:590-9. [PMID: 25336728 DOI: 10.1093/infdis/jiu481] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Dengue viruses (DENVs) are mosquito-borne flaviviruses that infect humans. The clinical presentation of DENV infection ranges from inapparent infection to dengue hemorrhagic fever and dengue shock syndrome. We analyzed samples from a pediatric dengue cohort study in Sri Lanka to explore whether antibody responses differentiated clinically apparent infections from clinically inapparent infections. In DENV-naive individuals exposed to primary DENV infections, we observed no difference in the quantity or quality of acquired antibodies between inapparent and apparent infections. Children who experienced primary infections had broad, serotype-cross-neutralizing antibody responses that narrowed in breadth to a single serotype over a 12-month period after infection. In DENV immune children who were experiencing a repeat infection, we observed a strong association between preexisting neutralizing antibodies and clinical outcome. Notably, children with preexisting monospecific neutralizing antibody responses were more likely to develop fever than children with cross-neutralizing responses. Preexisting DENV neutralizing antibodies are correlated with protection from dengue disease.
Collapse
Affiliation(s)
- Kizzmekia S Corbett
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill
| | - Leah Katzelnick
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill Centre of Pathogen Evolution, University of Cambridge, United Kingdom
| | | | | | | | - Aravinda M de Silva
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill
| |
Collapse
|
90
|
Li Z, Yang H, Yang J, Lin H, Wang W, Liu L, Zhao Y, Liu L, Zeng X, Yu Y, Li Y. Construction and preliminary investigation of a novel dengue serotype 4 chimeric virus using Japanese encephalitis vaccine strain SA14-14-2 as the backbone. Virus Res 2014; 191:10-20. [PMID: 25091563 DOI: 10.1016/j.virusres.2014.07.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/24/2014] [Accepted: 07/16/2014] [Indexed: 11/25/2022]
|
91
|
Wells TJ, Whitters D, Sevastsyanovich YR, Heath JN, Pravin J, Goodall M, Browning DF, O'Shea MK, Cranston A, De Soyza A, Cunningham AF, MacLennan CA, Henderson IR, Stockley RA. Increased severity of respiratory infections associated with elevated anti-LPS IgG2 which inhibits serum bactericidal killing. ACTA ACUST UNITED AC 2014; 211:1893-904. [PMID: 25113975 PMCID: PMC4144740 DOI: 10.1084/jem.20132444] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
An antibody directed against the O-antigen of Pseudomonas aeruginosa LPS can block complement-mediated bacterial killing and contributes to the severity of respiratory infection. Although specific antibody induced by pathogens or vaccines is a key component of protection against infectious threats, some viruses, such as dengue, induce antibody that enhances the development of infection. In contrast, antibody-dependent enhancement of bacterial infection is largely unrecognized. Here, we demonstrate that in a significant portion of patients with bronchiectasis and Pseudomonas aeruginosa lung infection, antibody can protect the bacterium from complement-mediated killing. Strains that resist antibody-induced, complement-mediated killing produce lipopolysaccharide containing O-antigen. The inhibition of antibody-mediated killing is caused by excess production of O-antigen–specific IgG2 antibodies. Depletion of IgG2 to O-antigen restores the ability of sera to kill strains with long-chain O-antigen. Patients with impaired serum-mediated killing of P. aeruginosa by IgG2 have poorer respiratory function than infected patients who do not produce inhibitory antibody. We suggest that excessive binding of IgG2 to O-antigen shields the bacterium from other antibodies that can induce complement-mediated killing of bacteria. As there is significant sharing of O-antigen structure between different Gram-negative bacteria, this IgG2-mediated impairment of killing may operate in other Gram-negative infections. These findings have marked implications for our understanding of protection generated by natural infection and for the design of vaccines, which should avoid inducing such blocking antibodies.
Collapse
Affiliation(s)
- Timothy J Wells
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Deborah Whitters
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Lung Investigation Unit, Queen Elizabeth Hospital, Birmingham B15 2TH, England, UK
| | - Yanina R Sevastsyanovich
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Jennifer N Heath
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - John Pravin
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Margaret Goodall
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Douglas F Browning
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Matthew K O'Shea
- The University of Oxford, The Jenner Institute, Oxford OX3 7DQ, England, UK
| | - Amy Cranston
- Sir William Leech Centre for Respiratory Research Newcastle upon Tyne Hospitals Trust, Newcastle NE7 7DN, England, UK
| | - Anthony De Soyza
- Institute of Cellular Medicine, Newcastle University and Adult Bronchiectasis service Freeman Hospital, Newcastle NE7 7DN, England, UK
| | - Adam F Cunningham
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Calman A MacLennan
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Ian R Henderson
- Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK Institute of Microbiology and Infection, School of Immunity and Infection, School Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, England, UK
| | - Robert A Stockley
- Lung Investigation Unit, Queen Elizabeth Hospital, Birmingham B15 2TH, England, UK
| |
Collapse
|
92
|
Abstract
Of the many pathogens that infect humans and animals, a large number use cells of the host organism as protected sites for replication. To reach the relevant intracellular compartments, they take advantage of the endocytosis machinery and exploit the network of endocytic organelles for penetration into the cytosol or as sites of replication. In this review, we discuss the endocytic entry processes used by viruses and bacteria and compare the strategies used by these dissimilar classes of pathogens.
Collapse
Affiliation(s)
- Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris F-75015, France; INSERM U604, Paris F-75015, France; and INRA, USC2020, Paris F-75015, France
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, 8093 Zurich, Switzerland
| |
Collapse
|
93
|
Richter MKS, da Silva Voorham JM, Torres Pedraza S, Hoornweg TE, van de Pol DPI, Rodenhuis-Zybert IA, Wilschut J, Smit JM. Immature dengue virus is infectious in human immature dendritic cells via interaction with the receptor molecule DC-SIGN. PLoS One 2014; 9:e98785. [PMID: 24886790 PMCID: PMC4041791 DOI: 10.1371/journal.pone.0098785] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/06/2014] [Indexed: 01/01/2023] Open
Abstract
Background Dengue Virus (DENV) is the most common mosquito-borne viral infection worldwide. Important target cells during DENV infection are macrophages, monocytes, and immature dendritic cells (imDCs). DENV-infected cells are known to secrete a large number of partially immature and fully immature particles alongside mature virions. Fully immature DENV particles are considered non-infectious, but antibodies have been shown to rescue their infectious properties. This suggests that immature DENV particles only contribute to the viral load observed in patients with a heterologous DENV re-infection. Methodology/Principal findings In this study, we re-evaluated the infectious properties of fully immature particles in absence and presence of anti-DENV human serum. We show that immature DENV is infectious in cells expressing DC-SIGN. Furthermore, we demonstrate that immature dendritic cells, in contrast to macrophage-like cells, do not support antibody-dependent enhancement of immature DENV. Conclusions/Significance Our data shows that immature DENV can infect imDCs through interaction with DC-SIGN, suggesting that immature and partially immature DENV particles may contribute to dengue pathogenesis during primary infection. Furthermore, since antibodies do not further stimulate DENV infectivity on imDCs we propose that macrophages/monocytes rather than imDCs contribute to the increased viral load observed during severe heterotypic DENV re-infections.
Collapse
Affiliation(s)
- Mareike K. S. Richter
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Júlia M. da Silva Voorham
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Silvia Torres Pedraza
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Grupo Immunovirología, Universidad de Antioquia, Medellín, Colombia
| | - Tabitha E. Hoornweg
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Denise P. I. van de Pol
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Wilschut
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jolanda M. Smit
- Department of Medical Microbiology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
94
|
Ng JKW, Zhang SL, Tan HC, Yan B, Maria Martinez Gomez J, Tan WY, Lam JH, Tan GKX, Ooi EE, Alonso S. First experimental in vivo model of enhanced dengue disease severity through maternally acquired heterotypic dengue antibodies. PLoS Pathog 2014; 10:e1004031. [PMID: 24699622 PMCID: PMC3974839 DOI: 10.1371/journal.ppat.1004031] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 02/11/2014] [Indexed: 11/18/2022] Open
Abstract
Dengue (DEN) represents the most serious arthropod-borne viral disease. DEN clinical manifestations range from mild febrile illness to life-threatening hemorrhage and vascular leakage. Early epidemiological observations reported that infants born to DEN-immune mothers were at greater risk to develop the severe forms of the disease upon infection with any serotype of dengue virus (DENV). From these observations emerged the hypothesis of antibody-dependent enhancement (ADE) of disease severity, whereby maternally acquired anti-DENV antibodies cross-react but fail to neutralize DENV particles, resulting in higher viremia that correlates with increased disease severity. Although in vitro and in vivo experimental set ups have indirectly supported the ADE hypothesis, direct experimental evidence has been missing. Furthermore, a recent epidemiological study has challenged the influence of maternal antibodies in disease outcome. Here we have developed a mouse model of ADE where DENV2 infection of young mice born to DENV1-immune mothers led to earlier death which correlated with higher viremia and increased vascular leakage compared to DENV2-infected mice born to dengue naïve mothers. In this ADE model we demonstrated the role of TNF-α in DEN-induced vascular leakage. Furthermore, upon infection with an attenuated DENV2 mutant strain, mice born to DENV1-immune mothers developed lethal disease accompanied by vascular leakage whereas infected mice born to dengue naïve mothers did no display any clinical manifestation. In vitro ELISA and ADE assays confirmed the cross-reactive and enhancing properties towards DENV2 of the serum from mice born to DENV1-immune mothers. Lastly, age-dependent susceptibility to disease enhancement was observed in mice born to DENV1-immune mothers, thus reproducing epidemiological observations. Overall, this work provides direct in vivo demonstration of the role of maternally acquired heterotypic dengue antibodies in the enhancement of dengue disease severity and offers a unique opportunity to further decipher the mechanisms involved.
Collapse
Affiliation(s)
- Jowin Kai Wei Ng
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | | | - Hwee Cheng Tan
- Progamme in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Benedict Yan
- Department of Pathology, National University Health System and National University of Singapore, Singapore
| | - Julia Maria Martinez Gomez
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | - Wei Yu Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | - Jian Hang Lam
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | - Grace Kai Xin Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
| | - Eng Eong Ooi
- Progamme in Emerging Infectious Diseases, Duke-NUS Graduate Medical School, Singapore
| | - Sylvie Alonso
- Department of Microbiology, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- Immunology Programme, Yong Loo Lin School of Medicine, Life Sciences Institute, National University of Singapore, Singapore
- * E-mail:
| |
Collapse
|
95
|
Liu P, Wang X, Itano MS, Neumann AK, de Silva AM, Jacobson K, Thompson NL. Low copy numbers of DC-SIGN in cell membrane microdomains: implications for structure and function. Traffic 2013; 15:179-96. [PMID: 24313910 DOI: 10.1111/tra.12138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 11/07/2013] [Accepted: 11/11/2013] [Indexed: 12/17/2022]
Abstract
Presently, there are few estimates of the number of molecules occupying membrane domains. Using a total internal reflection fluorescence microscopy (TIRFM) imaging approach, based on comparing the intensities of fluorescently labeled microdomains with those of single fluorophores, we measured the occupancy of DC-SIGN, a C-type lectin, in membrane microdomains. DC-SIGN or its mutants were labeled with primary monoclonal antibodies (mAbs) in either dendritic cells (DCs) or NIH3T3 cells, or expressed as GFP fusions in NIH3T3 cells. The number of DC-SIGN molecules per microdomain ranges from only a few to over 20, while microdomain dimensions range from the diffraction limit to > 1 µm. The largest fraction of microdomains, appearing at the diffraction limit, in either immature DCs or 3 T3 cells contains only 4-8 molecules of DC-SIGN, consistent with our preliminary super-resolution Blink microscopy estimates. We further show that these small assemblies are sufficient to bind and efficiently internalize a small (∼ 50 nm) pathogen, dengue virus, leading to infection of host cells.
Collapse
Affiliation(s)
- Ping Liu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7090, USA
| | | | | | | | | | | | | |
Collapse
|
96
|
Near-atomic resolution cryo-electron microscopic structure of dengue serotype 4 virus. J Virol 2013; 88:477-82. [PMID: 24155405 DOI: 10.1128/jvi.02641-13] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dengue virus (DENV), a mosquito-borne virus, is responsible for millions of cases of infections worldwide. There are four DENV serotypes (DENV1 to -4). After a primary DENV infection, the antibodies elicited confer lifetime protection against that DENV serotype. However, in a secondary infection with another serotype, the preexisting antibodies may cause antibody-dependent enhancement (ADE) of infection of macrophage cells, leading to the development of the more severe form of disease, dengue hemorrhagic fever. Thus, a safe vaccine should stimulate protection against all dengue serotypes simultaneously. To facilitate the development of a vaccine, good knowledge of different DENV serotype structures is crucial. Structures of DENV1 and DENV2 had been solved previously. Here we present a near-atomic resolution cryo-electron microscopy (cryo-EM) structure of mature DENV4. Comparison of the DENV4 structure with similar-resolution cryo-EM structures of DENV1 and DENV2 showed differences in surface charge distribution, which may explain their differences in binding to cellular receptors, such as heparin. Also, observed variations in amino acid residues involved in interactions between envelope and membrane proteins on the virus surface correlate with their ability to undergo structural changes at higher temperatures.
Collapse
|
97
|
The cytokine response of U937-derived macrophages infected through antibody-dependent enhancement of dengue virus disrupts cell apical-junction complexes and increases vascular permeability. J Virol 2013; 87:7486-501. [PMID: 23616663 DOI: 10.1128/jvi.00085-13] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Severe dengue (SD) is a life-threatening complication of dengue that includes vascular permeability syndrome (VPS) and respiratory distress. Secondary infections are considered a risk factor for developing SD, presumably through a mechanism called antibody-dependent enhancement (ADE). Despite extensive studies, the molecular bases of how ADE contributes to SD and VPS are largely unknown. This work compares the cytokine responses of differentiated U937 human monocytic cells infected directly with dengue virus (DENV) or in the presence of enhancing concentrations of a humanized monoclonal antibody recognizing protein E (ADE-DENV infection). Using a cytometric bead assay, ADE-DENV-infected cells were found to produce significantly higher levels of the proinflammatory cytokines interleukin 6 (IL-6), IL-12p70, and tumor necrosis factor alpha (TNF-α), as well as prostaglandin E2 (PGE2), than cells directly infected. The capacity of conditioned supernatants (conditioned medium [CM]) to disrupt tight junctions (TJs) in MDCK cell cultures was evaluated. Exposure of MDCK cell monolayers to CM collected from ADE-DENV-infected cells (ADE-CM) but not from cells infected directly led to a rapid loss of transepithelial electrical resistance (TER) and to delocalization and degradation of apical-junction complex proteins. Depletion of either TNF-α, IL-6, or IL-12p70 from CM from ADE-DENV-infected cells fully reverted the disrupting effect on TJs. Remarkably, mice injected intraperitoneally with ADE-CM showed increased vascular permeability in sera and lungs, as indicated by an Evans blue quantification assay. These results indicate that the cytokine response of U937-derived macrophages to ADE-DENV infection shows an increased capacity to disturb TJs, while results obtained with the mouse model suggest that such a response may be related to the vascular plasma leakage characteristic of SD.
Collapse
|