1
|
Sarker A, Rathore AS, Khalid MF, Gupta RD. Structure-guided affinity maturation of a single-chain variable fragment antibody against the Fu-bc epitope of the dengue virus envelope protein. J Biol Chem 2022; 298:101772. [PMID: 35218775 PMCID: PMC8956951 DOI: 10.1016/j.jbc.2022.101772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 11/05/2022] Open
Abstract
Dengue is one of the most dominant arthropod-borne viral diseases, infecting at least 390 million people every year throughout the world. Despite this, there is no effective treatment against dengue, and the only available vaccine has already been withdrawn owing to the significant adverse effects. Therefore, passive immunotherapy using monoclonal antibodies is now being sought as a therapeutic option. To date, many dengue monoclonal antibodies have been identified, most of which are serotype-specific, and only a few of which are cross-reactive. Furthermore, antibodies that cross-react within serotypes are weakly neutralizing and frequently induce antibody-dependent enhancement, which promotes viral entry and replication. Therefore, broadly neutralizing antibodies with no risk of antibody-dependent enhancement are required for the treatment of dengue. Here, we developed a single-chain variable fragment (scFv) antibody from an anti-fusion loop E53 antibody (PDB: 2IGF). We introduced previously predicted favorable complementarity-determining region (CDR) mutations into the gene encoding the scFv antibody for affinity maturation, and the resultant variants were tested in vitro against the highly conserved fusion and bc epitope of the dengue virus envelope protein. We show some of these scFv variants with two to three substitution mutations in three different CDRs possess affinity constants (KD) ranging from 20 to 200 nM. The scFv-mutant15, containing D31L, Y105W, and S227W substitutions, showed the lowest affinity constant, (KD = 24 ± 7 nM), approximately 100-fold lower than its parental construct. We propose that the scFv-derivative antibody may be a good candidate for the development of an effective and safe immunotherapy.
Collapse
Affiliation(s)
- Animesh Sarker
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, India
| | - Abhishek Singh Rathore
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, India
| | - Md Fahim Khalid
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, India
| | - Rinkoo Devi Gupta
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, India.
| |
Collapse
|
2
|
Pinto AK, Hassert M, Han X, Barker D, Carnelley T, Branche E, Steffen TL, Stone ET, Geerling E, Viramontes KM, Nykiforuk C, Toth D, Shresta S, Kodihalli S, Brien JD. The Ability of Zika virus Intravenous Immunoglobulin to Protect From or Enhance Zika Virus Disease. Front Immunol 2021; 12:717425. [PMID: 34552587 PMCID: PMC8450494 DOI: 10.3389/fimmu.2021.717425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022] Open
Abstract
The closely related flaviviruses, dengue and Zika, cause significant human disease throughout the world. While cross-reactive antibodies have been demonstrated to have the capacity to potentiate disease or mediate protection during flavivirus infection, the mechanisms responsible for this dichotomy are still poorly understood. To understand how the human polyclonal antibody response can protect against, and potentiate the disease in the context of dengue and Zika virus infection we used intravenous hyperimmunoglobulin (IVIG) preparations in a mouse model of the disease. Three IVIGs (ZIKV-IG, Control-Ig and Gamunex®) were evaluated for their ability to neutralize and/or enhance Zika, dengue 2 and 3 viruses in vitro. The balance between virus neutralization and enhancement provided by the in vitro neutralization data was used to predict the IVIG concentrations which could protect or enhance Zika, and dengue 2 disease in vivo. Using this approach, we were able to define the unique in vivo dynamics of complex polyclonal antibodies, allowing for both enhancement and protection from flavivirus infection. Our results provide a novel understanding of how polyclonal antibodies interact with viruses with implications for the use of polyclonal antibody therapeutics and the development and evaluation of the next generation flavivirus vaccines.
Collapse
Affiliation(s)
- Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Xiaobing Han
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | | | | | - Emilie Branche
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | - Tara L. Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - E. Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| | - Karla M. Viramontes
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - Derek Toth
- Emergent BioSolutions Canada Inc, Winnipeg, Canada
| | - Sujan Shresta
- La Jolla Institute for Immunology, Center for Infectious Disease and Vaccine Research, La Jolla, CA, United States
| | | | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St Louis, MO, United States
| |
Collapse
|
3
|
Earnest JT, Holmes AC, Basore K, Mack M, Fremont DH, Diamond MS. The mechanistic basis of protection by non-neutralizing anti-alphavirus antibodies. Cell Rep 2021; 35:108962. [PMID: 33826892 PMCID: PMC8055377 DOI: 10.1016/j.celrep.2021.108962] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 01/19/2021] [Accepted: 03/16/2021] [Indexed: 12/22/2022] Open
Abstract
Although neutralizing monoclonal antibodies (mAbs) against epitopes within the alphavirus E2 protein can protect against infection, the functional significance of non-neutralizing mAbs is poorly understood. Here, we evaluate the activity of 13 non-neutralizing mAbs against Mayaro virus (MAYV), an emerging arthritogenic alphavirus. These mAbs bind to the MAYV virion and surface of infected cells but fail to neutralize infection in cell culture. Mapping studies identify six mAb binding groups that localize to discrete epitopes within or adjacent to the A domain of the E2 glycoprotein. Remarkably, passive transfer of non-neutralizing mAbs protects against MAYV infection and disease in mice, and their efficacy requires Fc effector functions. Monocytes mediate the protection of non-neutralizing mAbs in vivo, as Fcγ-receptor-expressing myeloid cells facilitate the binding, uptake, and clearance of MAYV without antibody-dependent enhancement of infection. Humoral protection against alphaviruses likely reflects contributions from non-neutralizing antibodies through Fc-dependent mechanisms that accelerate viral clearance.
Collapse
Affiliation(s)
- James T Earnest
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Autumn C Holmes
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine Basore
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthias Mack
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine. St. Louis, MO 63110, USA.
| |
Collapse
|
4
|
Shukla R, Beesetti H, Brown JA, Ahuja R, Ramasamy V, Shanmugam RK, Poddar A, Batra G, Krammer F, Lim JK, Kale S, Lal AA, Swaminathan S, Khanna N. Dengue and Zika virus infections are enhanced by live attenuated dengue vaccine but not by recombinant DSV4 vaccine candidate in mouse models. EBioMedicine 2020; 60:102991. [PMID: 32949997 PMCID: PMC7501058 DOI: 10.1016/j.ebiom.2020.102991] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 08/03/2020] [Accepted: 08/20/2020] [Indexed: 01/21/2023] Open
Abstract
Background A tetravalent live attenuated dengue vaccine, Dengvaxia, sensitised naïve recipients to severe dengue illness upon a subsequent natural dengue infection and is suspected to be due to antibody-dependent enhancement (ADE). ADE has also been implicated in the severe neurological outcomes of Zika virus (ZIKV) infection. It has become evident that cross-reactive antibodies targeting the viral pre-membrane protein and fusion-loop epitope are ADE-competent. A pre-clinical tetravalent dengue sub-unit vaccine candidate, DSV4, eliminates these ADE-competent epitopes. Methods We compared protective efficacy and ADE-competence of murine polyclonal antibodies induced by DSV4, Dengvaxia and an ‘in house’ tetravalent mixture of all four laboratory DENV strains, TV DENV, using established mouse models. Findings DSV4-induced antibodies, known to be predominantly type-specific, provided significant protection against lethal DENV challenge, but did not promote ADE of either DENV or ZIKV infection in vivo. Antibodies elicited by Dengvaxia and TV DENV, which are predominantly cross-reactive, not only failed to offer protection against lethal DENV challenge, but also promoted ADE of both DENV and ZIKV infection in vivo. Interpretation Protective efficacy against DENV infection may be linked to the induction of neutralising antibodies which are type-specific rather than cross-reactive. Whole virus-based dengue vaccines may be associated with ADE risk, despite their potent virus-neutralising capacity. Vaccines designed to eliminate ADE-competent epitopes may help eliminate/minimise ADE risk. Funding This study was supported partly by ICGEB, India, the National Biopharma Mission, DBT, Government of India, Sun Pharmaceutical Industries Limited, India, and NIAID, NIH, USA.
Collapse
Affiliation(s)
- Rahul Shukla
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Hemalatha Beesetti
- Dengue Laboratory, Sun Pharmaceutical Industries Ltd., Gurugram, Haryana, India
| | - Julia A Brown
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Richa Ahuja
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Viswanathan Ramasamy
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Rajgokul K Shanmugam
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Ankur Poddar
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India
| | - Gaurav Batra
- Centre for Biodesign and Diagnostics, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, Faridabad, India
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jean K Lim
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Sachin Kale
- Dengue Laboratory, Sun Pharmaceutical Industries Ltd., Gurugram, Haryana, India
| | - Altaf A Lal
- Dengue Laboratory, Sun Pharmaceutical Industries Ltd., Gurugram, Haryana, India
| | - Sathyamangalam Swaminathan
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India.
| | - Navin Khanna
- Translational Health Group, Molecular Medicine Division, International Centre for Genetic Engineering & Biotechnology, New Delhi, India.
| |
Collapse
|
5
|
Maslow JN. Vaccines for emerging infectious diseases: Lessons from MERS coronavirus and Zika virus. Hum Vaccin Immunother 2017; 13:2918-2930. [PMID: 28846484 PMCID: PMC5718785 DOI: 10.1080/21645515.2017.1358325] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The past decade and a half has been characterized by numerous emerging infectious diseases. With each new threat, there has been a call for rapid vaccine development. Pathogens such as the Middle East Respiratory Syndrome coronavirus (MERS-CoV) and the Zika virus represent either new viral entities or viruses emergent in new geographic locales and characterized by novel complications. Both serve as paradigms for the global spread that can accompany new pathogens. In this paper, we review the epidemiology and pathogenesis of MERS-CoV and Zika virus with respect to vaccine development. The challenges in vaccine development and the approach to clinical trial design to test vaccine candidates for disease entities with a changing epidemiology are discussed.
Collapse
|
6
|
Dengue virus induces mitochondrial elongation through impairment of Drp1-triggered mitochondrial fission. Virology 2016; 500:149-160. [PMID: 27816895 DOI: 10.1016/j.virol.2016.10.022] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 02/08/2023]
Abstract
Mitochondria are highly dynamic organelles that undergo continuous cycles of fission and fusion to maintain essential cellular functions. An imbalance between these two processes can result in many pathophysiological outcomes. Dengue virus (DENV) interacts with cellular organelles, including mitochondria, to successfully replicate in cells. This study used live-cell imaging and found an increase in mitochondrial length and respiration during DENV infection. The level of mitochondrial fission protein, Dynamin-related protein 1 (Drp1), was decreased on mitochondria during DENV infection, as well as Drp1 phosphorylated on serine 616, which is important for mitochondrial fission. DENV proteins NS4b and NS3 were also associated with subcellular fractions of mitochondria. Induction of fission through uncoupling of mitochondria or overexpression of Drp1 wild-type and Drp1 with a phosphomimetic mutation (S616D) significantly reduced viral replication. These results demonstrate that DENV infection causes an imbalance in mitochondrial dynamics by inhibiting Drp1-triggered mitochondrial fission, which promotes viral replication.
Collapse
|
7
|
Cruz Hernández SIDL, Puerta-Guardo HN, Flores Aguilar H, González Mateos S, López Martinez I, Ortiz-Navarrete V, Ludert JE, Angel RMD. Primary dengue virus infections induce differential cytokine production in Mexican patients. Mem Inst Oswaldo Cruz 2016; 111:161-7. [PMID: 27008374 PMCID: PMC4804498 DOI: 10.1590/0074-02760150359] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/12/2016] [Indexed: 12/17/2022] Open
Abstract
Severe dengue pathogenesis is not fully understood, but high levels of
proinflammatory cytokines have been associated with dengue disease severity. In this
study, the cytokine levels in 171 sera from Mexican patients with primary dengue
fever (DF) and dengue haemorrhagic fever (DHF) from dengue virus (DENV) 1 (n = 116)
or 2 (n = 55) were compared. DF and DHF were defined according to the patient’s
clinical condition, the primary infections as indicated by IgG enzymatic immunoassay
negative results, and the infecting serotype as assessed by real-time reverse
transcription-polymerase chain reaction. Samples were analysed for circulating levels
of interleukin (IL)-12p70, interferon (IFN)-γ, tumour necrosis factor (TNF)-α, IL-6,
and IL-8 using a commercial cytometric bead array. Significantly higher IFN-γ levels
were found in patients with DHF than those with DF. However, significantly higher
IL-12p70, TNF-α, and IL-6 levels were associated with DHF only in patients who were
infected with DENV2 but not with DENV1. Moreover, patients with DF who were infected
with DENV1 showed higher levels of IL-12p70, TNF-α, and IL-6 than patients with DHF
early after-fever onset. The IL-8 levels were similar in all cases regardless of the
clinical condition or infection serotype. These results suggest that the association
between high proinflammatory cytokine levels and dengue disease severity does not
always stand, and it once again highlights the complex nature of DHF
pathogenesis.
Collapse
Affiliation(s)
| | - Henry Nelson Puerta-Guardo
- Departamento de Infectómica y Patogénesis Molecular, Instituto Politécnico Nacional, Centro de Investigación y Estudios Avanzados, México, DF, México
| | - Hilario Flores Aguilar
- Departamento de Inmunología e Inmunogenética, Instituto de Diagnóstico y Referencia Epidemiológicos, México, DF, México
| | - Silvia González Mateos
- Departamento de Virología, Instituto de Diagnóstico y Referencia Epidemiológicos, México, DF, México
| | - Irma López Martinez
- Departamento de Virología, Instituto de Diagnóstico y Referencia Epidemiológicos, México, DF, México
| | - Vianney Ortiz-Navarrete
- Departamento de Biomedicina, Instituto Politécnico Nacional, Centro de Investigación y de Estudios Avanzados, México, DF, México
| | - Juan E Ludert
- Departamento de Infectómica y Patogénesis Molecular, Instituto Politécnico Nacional, Centro de Investigación y Estudios Avanzados, México, DF, México
| | - Rosa María del Angel
- Departamento de Infectómica y Patogénesis Molecular, Instituto Politécnico Nacional, Centro de Investigación y Estudios Avanzados, México, DF, México
| |
Collapse
|
8
|
Abstract
Introduction: Zika virus (ZIKV) has emerged in dengue (DENV) endemic areas, where
these two related flaviviruses continue to co-circulate. DENV is a complex of
four serotypes and infections can progress to severe disease. It is thought that
this is mediated by antibody dependent enhancement (ADE) whereby antibodies from
a primary DENV infection are incapable of neutralizing heterologous DENV
infections with another serotype. ADE has been demonstrated among other members
of the Flavivirus group. Methods: We utilize an in vitro ADE assay developed for DENV to determine whether
ZIKV is enhanced by a commonly available DENV serotype 2-derived monoclonal
antibody (4G2). Results: We show that ZIKV infection in vitro is enhanced in the presence of the
4G2 mAb. Discussion: Our results demonstrate that ADE between ZIKV and DENV is possible
and that the 4G2 antibody is a useful tool for the effects of pre-existing
anti-DENV antibodies during ZIKV infections.
Collapse
Affiliation(s)
- Anu Susan Charles
- Pathobiological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | | |
Collapse
|
9
|
Dejnirattisai W, Supasa P, Wongwiwat W, Rouvinski A, Barba-Spaeth G, Duangchinda T, Sakuntabhai A, Cao-Lormeau VM, Malasit P, Rey FA, Mongkolsapaya J, Screaton GR. Dengue virus sero-cross-reactivity drives antibody-dependent enhancement of infection with zika virus. Nat Immunol 2016; 17:1102-8. [PMID: 27339099 PMCID: PMC4994874 DOI: 10.1038/ni.3515] [Citation(s) in RCA: 689] [Impact Index Per Article: 76.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/14/2016] [Indexed: 12/12/2022]
Abstract
Zika virus (ZIKV) was discovered in 1947 and was thought to lead to relatively mild disease. The recent explosive outbreak of ZIKV in South America has led to widespread concern with reports of neurological sequelae ranging from Guillain Barré syndrome to microcephaly. ZIKV infection has occurred in areas previously exposed to dengue, a flavivirus closely related to ZIKV. Here we investigate the serological crossreaction between the two viruses. Dengue immune plasma substantially crossreacted with ZIKV and could drive antibody-dependent enhancement of ZIKV infection. Using a panel of human anti-dengue monoclonal antibodies we showed that most antibodies reacting to dengue envelope protein also reacted to ZIKV. Antibodies to linear epitopes including the immunodominant fusion loop epitope while able to bind ZIKV could not neutralize the virus but instead promoted ADE. These data indicate that dengue immunity may drive higher ZIKV replication and have clear implications for disease pathogenesis and future ZIKV and dengue vaccine programs.
Collapse
Affiliation(s)
- Wanwisa Dejnirattisai
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Piyada Supasa
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK.,Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand.,Graduate Program in Immunology, Department of Immunology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wiyada Wongwiwat
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| | - Alexander Rouvinski
- Institut Pasteur, Département de Virologie, Unité de Virologie Structurale, Paris, France.,CNRS UMR 3569 Virologie, Paris, France
| | - Giovanna Barba-Spaeth
- Institut Pasteur, Département de Virologie, Unité de Virologie Structurale, Paris, France.,CNRS UMR 3569 Virologie, Paris, France
| | - Thaneeya Duangchinda
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Anavaj Sakuntabhai
- Institut Pasteur, Functional Genetics of Infectious Diseases Unit, Paris, France.,CNRS URA3012, Paris, France
| | - Van-Mai Cao-Lormeau
- Unit of Emerging Infectious Diseases, Institut Louis Malardé, Papeete, Tahiti, French Polynesia
| | - Prida Malasit
- Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand.,Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Felix A Rey
- Institut Pasteur, Département de Virologie, Unité de Virologie Structurale, Paris, France.,CNRS UMR 3569 Virologie, Paris, France
| | - Juthathip Mongkolsapaya
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK.,Dengue Hemorrhagic Fever Research Unit, Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol University, Bangkok, Thailand
| | - Gavin R Screaton
- Division of Immunology and Inflammation, Department of Medicine, Hammersmith Campus, Imperial College London, UK
| |
Collapse
|
10
|
Defining New Therapeutics Using a More Immunocompetent Mouse Model of Antibody-Enhanced Dengue Virus Infection. mBio 2015; 6:e01316-15. [PMID: 26374123 PMCID: PMC4600115 DOI: 10.1128/mbio.01316-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
With over 3.5 billion people at risk and approximately 390 million human infections per year, dengue virus (DENV) disease strains health care resources worldwide. Previously, we and others established models for DENV pathogenesis in mice that completely lack subunits of the receptors (Ifnar and Ifngr) for type I and type II interferon (IFN) signaling; however, the utility of these models is limited by the pleotropic effect of these cytokines on innate and adaptive immune system development and function. Here, we demonstrate that the specific deletion of Ifnar expression on subsets of murine myeloid cells (LysM Cre+Ifnarflox/flox [denoted as Ifnarf/f herein]) resulted in enhanced DENV replication in vivo. The administration of subneutralizing amounts of cross-reactive anti-DENV monoclonal antibodies to LysM Cre+Ifnarf/f mice prior to infection with DENV serotype 2 or 3 resulted in antibody-dependent enhancement (ADE) of infection with many of the characteristics associated with severe DENV disease in humans, including plasma leakage, hypercytokinemia, liver injury, hemoconcentration, and thrombocytopenia. Notably, the pathogenesis of severe DENV-2 or DENV-3 infection in LysM Cre+Ifnarf/f mice was blocked by pre- or postexposure administration of a bispecific dual-affinity retargeting molecule (DART) or an optimized RIG-I receptor agonist that stimulates innate immune responses. Our findings establish a more immunocompetent animal model of ADE of infection with multiple DENV serotypes in which disease is inhibited by treatment with broad-spectrum antibody derivatives or innate immune stimulatory agents. Although dengue virus (DENV) infects hundreds of millions of people annually and results in morbidity and mortality on a global scale, there are no approved antiviral treatments or vaccines. Part of the difficulty in evaluating therapeutic candidates is the lack of small animal models that are permissive to DENV and recapitulate the clinical features of severe human disease. Using animals lacking the type I interferon receptor only on myeloid cell subsets, we developed a more immunocompetent mouse model of severe DENV infection with characteristics of the human disease, including vascular leakage, hemoconcentration, thrombocytopenia, and liver injury. Using this model, we demonstrate that pathogenesis by two different DENV serotypes is inhibited by therapeutic administration of a genetically modified antibody or a RIG-I receptor agonist that stimulates innate immunity.
Collapse
|
11
|
Bennett KM, Gorham RD, Gusti V, Trinh L, Morikis D, Lo DD. Hybrid flagellin as a T cell independent vaccine scaffold. BMC Biotechnol 2015; 15:71. [PMID: 26265529 PMCID: PMC4534063 DOI: 10.1186/s12896-015-0194-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 07/29/2015] [Indexed: 12/12/2022] Open
Abstract
Background To extend the potency of vaccines against infectious diseases, vaccines should be able to exploit multiple arms of the immune system. One component of the immune system that is under-used in vaccine design is the subset of B cells known to be capable of responding to repetitive antigenic epitopes and differentiate into plasma cells even in the absence of T cell help (T-independent, TI). Results To target vaccine responses from T-independent B cells, we reengineered a bacterial Flagellin (FliC) by replacing its exposed D3 domain with a viral envelope protein from Dengue virus (DENV2). The resulting hybrid FliC protein (hFliC) was able to form stable filaments decorated with conformationally intact DENV2 envelope domains. These filaments were not only capable of inducing a T cell-dependent (TD) humoral antibody response, but also significant IgM and IgG3 antibody response in a helper T cell repertoire-restricted transgenic mouse model. Conclusions Our results provide proof-of-principle demonstration that a reengineered hybrid FliC could be used as a platform for polymeric subunit vaccines, enhancing T cell-dependent and possibly inducing T-independent antibody responses from B-1 B cells as well. Electronic supplementary material The online version of this article (doi:10.1186/s12896-015-0194-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kaila M Bennett
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA. .,Bioengineering Interdepartmental Graduate Program, California, USA.
| | - Ronald D Gorham
- Department of Bioengineering, University of California Riverside, California, 92521, USA.
| | - Veronica Gusti
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| | - Lien Trinh
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| | - Dimitrios Morikis
- Department of Bioengineering, University of California Riverside, California, 92521, USA.
| | - David D Lo
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, California, 92521, USA.
| |
Collapse
|
12
|
Seanpong P, Srisaowakarn C, Thammaporn A, Leardkamolkarn V, Kumkate S. Different Responses in MMP/TIMP Expression of U937 and HepG2 Cells to Dengue Virus Infection. Jpn J Infect Dis 2015; 68:221-229. [PMID: 25672410 DOI: 10.7883/yoken.jjid.2013.481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Disease severities following dengue virus (DV) infection are the result of increased vascular permeability leading to hypovolemic shock. Matrix metalloproteinases (MMPs) are believed to play a key role in promoting such severities. A previous study reported that supernatants of DV-infected dendritic cells (DCs), which contained high levels of MMP-2 and MMP-9, induced vascular leakage in a mouse model. In the present study, we investigated whether hepatocytes (HepG2) and monocytes (U937) could be additional sources of MMPs during DV infection. HepG2 and U937 cells were exposed to DV serotype 2 strain 16681. The secretion of MMP-2 and MMP-9 was detected using gelatin zymography. We found that DV infection in the HepG2 cells promoted MMP-2 production while that in the U937 cells promoted MMP-9 production. Semi-quantitative RT-PCR results also confirmed that DV infection in the HepG2 cells up-regulated the expression of MMP-2 mRNA, whereas that in the U937 cells enhanced the expression of MMP-9 mRNA. We monitored the expression of endogenous TIMP-1 and TIMP-2. DV infection induced TIMP-1 expression in the U937 cells. However, lower expression of TIMP-2 was observed in the infected HepG2 cells. We believed that following DV infection, monocytes and hepatocytes can act as MMP-9 and MMP-2 producers, respectively. Their responses could be attributed to the disturbance of TIMP expression by DV in different cell types.
Collapse
|
13
|
De La Cruz Hernández SI, Puerta-Guardo H, Flores-Aguilar H, González-Mateos S, López-Martinez I, Ortiz-Navarrete V, Ludert JE, Del Angel RM. A strong interferon response correlates with a milder dengue clinical condition. J Clin Virol 2014; 60:196-9. [PMID: 24793964 DOI: 10.1016/j.jcv.2014.04.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 11/28/2013] [Accepted: 04/02/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Type 1 interferon (IFNα/β) has a significant role in establishing protection against virus infections. It has been well documented by in vitro studies that dengue virus (DENV) activates a robust IFNα/β response. However, DENV also induces a down-regulation of the JAK/STAT pathway, inhibiting the induction of interferon regulated genes. As a consequence, the role played by the IFN type 1 response in the protection of dengue patients is not fully understood. OBJECTIVE To compare IFN-α levels in dengue patients with dengue fever (DF) or dengue hemorrhagic fever (DHF) undergoing primary or secondary infections. STUDY DESIGN Two hundred and four serum samples were analyzed for IFN-α level by cytometric bead array. Patients' clinical condition was assigned following the WHO 1997 criteria and specific IgG and IgM antibodies were measured using commercial assays to determine primary and secondary infections. The infecting serotype was determined by qRT-PCR. RESULTS AND CONCLUSION The IFN-α levels were found significantly higher in DF than DHF patients irrespective of the infecting serotype (DENV1 or 2), and were found to decline rapidly at day 3 after fever onset. For DENV2 infections, higher IFN-α level was found during primary than secondary infections. These results suggest that an early strong interferon response correlates with a better clinical condition.
Collapse
Affiliation(s)
- Sergio Isaac De La Cruz Hernández
- Department of Virology, Institute for Diagnosis and Epidemiological Reference (InDRE), Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico; Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Henry Puerta-Guardo
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Hilario Flores-Aguilar
- Department of Inmunology and Inmunogenetics-InDRE, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Silvia González-Mateos
- Department of Virology, Institute for Diagnosis and Epidemiological Reference (InDRE), Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Irma López-Martinez
- Department of Virology, Institute for Diagnosis and Epidemiological Reference (InDRE), Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Vianney Ortiz-Navarrete
- Department of Biomedicine, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico
| | - Juan E Ludert
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico.
| | - Rosa María Del Angel
- Department of Infectomics and Molecular Pathogenesis, Center for Research and Advanced Studies (CINVESTAV-IPN), Mexico D.F., Mexico.
| |
Collapse
|
14
|
Simulations to compare efficacies of tetravalent dengue vaccines and mosquito vector control. Epidemiol Infect 2013; 142:1245-58. [PMID: 23925059 PMCID: PMC4045171 DOI: 10.1017/s0950268813001866] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SUMMARY Infection with dengue, the most prevalent mosquito-borne virus, manifests as dengue fever (DF) or the more fatal dengue haemorrhagic fever (DHF). DHF occurs mainly when an individual who has acquired antibodies to one serotype is inoculated with another serotype. It was reported that mosquito control may have increased the incidence of DF and DHF due to age-dependency in manifesting these illnesses or an immunological mechanism. Tetravalent dengue vaccine is currently being tested in clinical trials. However, seroconversions to all four serotypes were achieved only after three doses. Therefore, vaccines may predispose vaccinees to the risk of developing DHF in future infections. This study employed an individual-based computer simulation, to emulate mosquito control and vaccination, incorporating seroconversion rates reported from actual clinical trials. It was found that mosquito control alone would have increased incidence of DF and DHF in areas of high mosquito density. A vaccination programme with very high coverage, even with a vaccine of suboptimal seroconversion rates, attenuated possible surges in the incidence of DF and DHF which would have been caused by insufficient reduction in mosquito abundance. DHF cases attributable to vaccine-derived enhancement were fewer than DHF cases prevented by a vaccine with considerably high (although not perfect) seroconversion rates. These predictions may justify vaccination programmes, at least in areas of high mosquito abundance. In such areas, mosquito control programmes should be conducted only after the vaccination programme with a high coverage has been initiated.
Collapse
|
15
|
Abstract
Family Flaviviridae genus flavivirus contains numerous pathogenic viruses such as Japanese encephalitis virus, dengue virus, West Nile virus, etc, which cause public health problems in the world. Since many mammals and birds can act as amplifying hosts and reservoir hosts in nature and those viruses are transmitted by haematophagous mosquitoes or ticks, those viruses could not be eradicated from the nature. In the recent few decades, the viral replication mechanism and the ultrastructure of viral proteins as well as the viral immune evasion mechanism have been elucidated extensively, leading to develop novel types of antivirals and vaccines. In this review, the flavivirus nature and epidemiology, replication mechanism, immune response and immune evasion, and antivirals and vaccines against flaviviruses were described.
Collapse
Affiliation(s)
- Tomohiro Ishikawa
- Department of Microbiology, Dokkyo Medical University School of Medicine.
| | | |
Collapse
|
16
|
Rodenhuis-Zybert IA, van der Schaar HM, da Silva Voorham JM, van der Ende-Metselaar H, Lei HY, Wilschut J, Smit JM. Immature dengue virus: a veiled pathogen? PLoS Pathog 2010; 6:e1000718. [PMID: 20062797 PMCID: PMC2798752 DOI: 10.1371/journal.ppat.1000718] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 12/10/2009] [Indexed: 12/19/2022] Open
Abstract
Cells infected with dengue virus release a high proportion of immature prM-containing virions. In accordance, substantial levels of prM antibodies are found in sera of infected humans. Furthermore, it has been recently described that the rates of prM antibody responses are significantly higher in patients with secondary infection compared to those with primary infection. This suggests that immature dengue virus may play a role in disease pathogenesis. Interestingly, however, numerous functional studies have revealed that immature particles lack the ability to infect cells. In this report, we show that fully immature dengue particles become highly infectious upon interaction with prM antibodies. We demonstrate that prM antibodies facilitate efficient binding and cell entry of immature particles into Fc-receptor-expressing cells. In addition, enzymatic activity of furin is critical to render the internalized immature virus infectious. Together, these data suggest that during a secondary infection or primary infection of infants born to dengue-immune mothers, immature particles have the potential to be highly infectious and hence may contribute to the development of severe disease.
Collapse
Affiliation(s)
- Izabela A. Rodenhuis-Zybert
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Hilde M. van der Schaar
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Júlia M. da Silva Voorham
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Heidi van der Ende-Metselaar
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Huan-Yao Lei
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jan Wilschut
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | - Jolanda M. Smit
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
17
|
Puerta-Guardo H, Mosso C, Medina F, Liprandi F, Ludert JE, del Angel RM. Antibody-dependent enhancement of dengue virus infection in U937 cells requires cholesterol-rich membrane microdomains. J Gen Virol 2009; 91:394-403. [DOI: 10.1099/vir.0.015420-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
18
|
Structural basis for the preferential recognition of immature flaviviruses by a fusion-loop antibody. EMBO J 2009; 28:3269-76. [PMID: 19713934 DOI: 10.1038/emboj.2009.245] [Citation(s) in RCA: 179] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Accepted: 07/30/2009] [Indexed: 11/08/2022] Open
Abstract
Flaviviruses are a group of human pathogens causing severe encephalitic or hemorrhagic diseases that include West Nile, dengue and yellow fever viruses. Here, using X-ray crystallography we have defined the structure of the flavivirus cross-reactive antibody E53 that engages the highly conserved fusion loop of the West Nile virus envelope glycoprotein. Using cryo-electron microscopy, we also determined that E53 Fab binds preferentially to spikes in noninfectious, immature flavivirions but is unable to bind significantly to mature virions, consistent with the limited solvent exposure of the epitope. We conclude that the neutralizing impact of E53 and likely similar fusion-loop-specific antibodies depends on its binding to the frequently observed immature component of flavivirus particles. Our results elucidate how fusion-loop antibodies, which comprise a significant fraction of the humoral response against flaviviruses, can function to control infection without appreciably recognizing mature virions. As these highly cross-reactive antibodies are often weakly neutralizing they also may contribute to antibody-dependent enhancement and flavi virus pathogenesis thereby complicating development of safe and effective vaccines.
Collapse
|
19
|
Abstract
Dengue viruses (DENV) are the most important human arboviral pathogens. Transmission in tropical and subtropical regions of the world includes a sylvatic, enzootic cycle between nonhuman primates and arboreal mosquitoes of the genus Aedes, and an urban, endemic/epidemic cycle principally between Aedes aegypti, a mosquito that exploits peridomestic water containers as its larval habitats, and human reservoir hosts that are preferred for blood feeding. Genetic studies suggest that all four serotypes of endemic/epidemic DENV evolved independently from ancestral, sylvatic viruses and subsequently became both ecologically and evolutionarily distinct. The independent evolution of these four serotypes was accompanied by the expansion of the sylvatic progenitors' host range in Asia to new vectors and hosts, which probably occurred gradually over a period of several hundred years. Although many emerging viral pathogens adapt to human replication and transmission, the available evidence indicates that adaptation to humans is probably not a necessary component of sylvatic DENV emergence. These findings imply that the sylvatic DENV cycles in Asia and West Africa will remain a potential source of re-emergence. Sustained urban vector control programs and/or human vaccination will be required to control DEN because the enzootic vectors and primate reservoir hosts are not amenable to interventions.
Collapse
Affiliation(s)
- Nikos Vasilakis
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555-0609, USA
| | | |
Collapse
|
20
|
Oishi K, Saito M, Mapua CA, Natividad FF. Dengue illness: clinical features and pathogenesis. J Infect Chemother 2007; 13:125-33. [PMID: 17593497 DOI: 10.1007/s10156-007-0516-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Indexed: 10/23/2022]
Abstract
The incidence and geographical distribution of dengue has gradually increased during the past decade. This review is an update on dengue virus infections, based on our clinical and laboratory experiences in the Philippines and on other relevant literature. The differential diagnosis of this disease is discussed, especially for use by clinicians where dengue is not endemic. The complex pathogenesis of thrombocytopenia and increased vascular permeability in dengue illness is also discussed. Our recent data suggest that platelet-associated immunoglobulins involving anti-dengue virus activity play a pivotal role in the development of dengue hemorrhagic fever (DHF), as well as thrombocytopenia in secondary dengue virus infections. Further elucidation is needed on the involvement of platelet-associated immunoglobulins on the molecular mechanisms of thrombocytopenia and the increased vascular permeability.
Collapse
Affiliation(s)
- Kazunori Oishi
- Department of Special Pathogens, International Research Center for Infectious Diseases, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | | | | | | |
Collapse
|
21
|
Shresta S, Sharar KL, Prigozhin DM, Beatty PR, Harris E. Murine model for dengue virus-induced lethal disease with increased vascular permeability. J Virol 2006; 80:10208-17. [PMID: 17005698 PMCID: PMC1617308 DOI: 10.1128/jvi.00062-06] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Lack of an appropriate animal model for dengue virus (DEN), which causes dengue fever and dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS), has impeded characterization of the mechanisms underlying the disease pathogenesis. The cardinal feature of DHF/DSS, the severe form of DEN infection, is increased vascular permeability. To develop a murine model that is more relevant to DHF/DSS, a novel DEN strain, D2S10, was generated by alternately passaging a non-mouse-adapted DEN strain between mosquito cells and mice, thereby mimicking the natural transmission cycle of the virus between mosquitoes and humans. After infection with D2S10, mice lacking interferon receptors died early without manifesting signs of paralysis, carried infectious virus in both non-neuronal and neuronal tissues, and exhibited signs of increased vascular permeability. In contrast, mice infected with the parental DEN strain developed paralysis at late times after infection, contained detectable levels of virus only in the central nervous system, and displayed normal vascular permeability. In the mice infected with D2S10, but not the parental DEN strain, significant levels of serum tumor necrosis factor alpha (TNF-alpha) were produced, and the neutralization of TNF-alpha activity prevented early death of D2S10-infected mice. Sequence analysis comparing D2S10 to its parental strain implicated a conserved region of amino acid residues in the envelope protein as a possible source for the D2S10 phenotype. These results demonstrate that D2S10 causes a more relevant disease in mice and that TNF-alpha may be one of several key mediators of severe DEN-induced disease in mice. This report represents a significant advance in animal models for severe DEN disease, and it begins to provide mechanistic insights into DEN-induced disease in vivo.
Collapse
Affiliation(s)
- Sujan Shresta
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
22
|
Modis Y, Ogata S, Clements D, Harrison SC. Variable surface epitopes in the crystal structure of dengue virus type 3 envelope glycoprotein. J Virol 2005; 79:1223-31. [PMID: 15613349 PMCID: PMC538574 DOI: 10.1128/jvi.79.2.1223-1231.2005] [Citation(s) in RCA: 325] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dengue virus is an emerging global health threat. The major envelope glycoprotein, E, mediates viral attachment and entry by membrane fusion. Antibodies that bind but fail to neutralize noncognate serotypes enhance infection. We have determined the crystal structure of a soluble fragment of the envelope glycoprotein E from dengue virus type 3. The structure closely resembles those of E proteins from dengue type 2 and tick-borne encephalitis viruses. Serotype-specific neutralization escape mutants in dengue virus E proteins are all located on a surface of domain III, which has been implicated in receptor binding. While antibodies against epitopes in domain I are nonneutralizing in dengue virus, there are neutralizing antibodies that recognize serotype-conserved epitopes in domain II. The mechanism of neutralization for these antibodies is probably inhibition of membrane fusion. Our structure shows that neighboring glycans on the viral surface are spaced widely enough (at least 32 A) that they can interact with multiple carbohydrate recognition domains on oligomeric lectins such as DC-SIGN, ensuring maximum affinity for these putative receptors.
Collapse
Affiliation(s)
- Yorgo Modis
- Children's Hospital, Enders 673, 320 Longwood Ave., Boston, MA 02115, USA
| | | | | | | |
Collapse
|
23
|
Diamond MS, Sitati EM, Friend LD, Higgs S, Shrestha B, Engle M. A critical role for induced IgM in the protection against West Nile virus infection. ACTA ACUST UNITED AC 2003; 198:1853-62. [PMID: 14662909 PMCID: PMC2194144 DOI: 10.1084/jem.20031223] [Citation(s) in RCA: 230] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In humans, the elderly and immunocompromised are at greatest risk for disseminated West Nile virus (WNV) infection, yet the immunologic basis for this remains unclear. We demonstrated previously that B cells and IgG contributed to the defense against disseminated WNV infection (Diamond, M.S., B. Shrestha, A. Marri, D. Mahan, and M. Engle. 2003. J. Virol. 77:2578–2586). In this paper, we addressed the function of IgM in controlling WNV infection. C57BL/6J mice (sIgM−/−) that were deficient in the production of secreted IgM but capable of expressing surface IgM and secreting other immunoglobulin isotypes were vulnerable to lethal infection, even after inoculation with low doses of WNV. Within 96 h, markedly higher levels of infectious virus were detected in the serum of sIgM−/− mice compared with wild-type mice. The enhanced viremia correlated with higher WNV burdens in the central nervous system, and was also associated with a blunted anti-WNV IgG response. Passive transfer of polyclonal anti-WNV IgM or IgG protected sIgM−/− mice against mortality, although administration of comparable amounts of a nonneutralizing monoclonal anti-WNV IgM provided no protection. In a prospective analysis, a low titer of anti-WNV IgM antibodies at day 4 uniformly predicted mortality in wild-type mice. Thus, the induction of a specific, neutralizing IgM response early in the course of WNV infection limits viremia and dissemination into the central nervous system, and protects against lethal infection.
Collapse
Affiliation(s)
- Michael S Diamond
- Washington University School of Medicine, 660 S. Euclid Ave, Box 8051, St Louis, MO 63110, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Shrestha B, Gottlieb D, Diamond MS. Infection and injury of neurons by West Nile encephalitis virus. J Virol 2003; 77:13203-13. [PMID: 14645577 PMCID: PMC296085 DOI: 10.1128/jvi.77.24.13203-13213.2003] [Citation(s) in RCA: 192] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2003] [Accepted: 09/12/2003] [Indexed: 11/20/2022] Open
Abstract
West Nile virus (WNV) infects neurons and leads to encephalitis, paralysis, and death in humans, animals, and birds. We investigated the mechanism by which neuronal injury occurs after WNV infection. Neurons in the anterior horn of the spinal cords of paralyzed mice exhibited a high degree of WNV infection, leukocyte infiltration, and degeneration. Because it was difficult to distinguish whether neuronal injury was caused by viral infection or by the immune system response, a novel tissue culture model for WNV infection was established in neurons derived from embryonic stem (ES) cells. Undifferentiated ES cells were relatively resistant to WNV infection. After differentiation, ES cells expressed neural antigens, acquired a neuronal phenotype, and became permissive for WNV infection. Within 48 h of exposure to an exceedingly low multiplicity of infection (5 x 10(-4)), 50% of ES cell-derived neurons became infected, producing nearly 10(7) PFU of infectious virus per ml, and began to die by an apoptotic mechanism. The establishment of a tractable virus infection model in ES cell-derived neurons facilitates the study of the molecular basis of neurotropism and the mechanisms of viral and immune-mediated neuronal injury after infection by WNV or other neurotropic pathogens.
Collapse
Affiliation(s)
- Bimmi Shrestha
- Departments of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
25
|
McCown M, Diamond MS, Pekosz A. The utility of siRNA transcripts produced by RNA polymerase i in down regulating viral gene expression and replication of negative- and positive-strand RNA viruses. Virology 2003; 313:514-24. [PMID: 12954218 DOI: 10.1016/s0042-6822(03)00341-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Short interfering double-stranded RNAs (siRNAs) expressed under the control of an RNA polymerase I promoter system were used to target gene expression of influenza A and West Nile virus. Decreased RNA and protein expression was induced in a sequence-specific manner-reducing sequence complementarity from 21 to 17 nucleotides abrogated the siRNA effect. Reduced M(2) expression resulted in a decrease in total and infectious influenza A virus production. WNV protein expression, genomic RNA, and infectious virus production were all dramatically reduced by siRNAs targeting two distinct viral sequences. The data demonstrate the utility of plasmid-driven siRNAs in regulating the expression of single viral genes, global viral gene expression, as a potential antiviral treatment, and as a genetic tool for viruses whose genomes are difficult to manipulate.
Collapse
Affiliation(s)
- Matthew McCown
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110-1093, USA
| | | | | |
Collapse
|
26
|
Abstract
In general, virus-specific antibodies are considered antiviral and play an important role in the control of virus infections in a number of ways. However, in some instances, the presence of specific antibodies can be beneficial to the virus. This activity is known as antibody-dependent enhancement (ADE) of virus infection. The ADE of virus infection is a phenomenon in which virus-specific antibodies enhance the entry of virus, and in some cases the replication of virus, into monocytes/macrophages and granulocytic cells through interaction with Fc and/or complement receptors. This phenomenon has been reported in vitro and in vivo for viruses representing numerous families and genera of public health and veterinary importance. These viruses share some common features such as preferential replication in macrophages, ability to establish persistence, and antigenic diversity. For some viruses, ADE of infection has become a great concern to disease control by vaccination. Consequently, numerous approaches have been made to the development of vaccines with minimum or no risk for ADE. Identification of viral epitopes associated with ADE or neutralization is important for this purpose. In addition, clear understanding of the cellular events after virus entry through ADE has become crucial for developing efficient intervention. However, the mechanisms of ADE still remain to be better understood.
Collapse
Affiliation(s)
- Sol M Cancel Tirado
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, Iowa, USA
| | | |
Collapse
|
27
|
Abstract
After a virus infects an animal, antiviral responses are generated that attempt to prevent dissemination. Interferons, antibody, complement, T and natural killer cells all contribute to the control and eradication of viral infections. Most flaviviruses, with the exception of some of the encephalitic viruses, cause acute disease and do not establish persistent infection. The outcome of flavivirus infection in an animal is determined by a balance between the speed of viral replication and spread, and the immune system response. Although many of the mechanistic details require further elucidation, flaviviruses have evolved specific tactics to evade the innate and adaptive immune response. A more thorough understanding of these principles could lead to improved models for viral pathogenesis and to strategies for the development of novel antiviral agents.
Collapse
Affiliation(s)
- Michael S Diamond
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri 63110, United States of America.
| |
Collapse
|
28
|
Cologna R, Rico-Hesse R. American genotype structures decrease dengue virus output from human monocytes and dendritic cells. J Virol 2003; 77:3929-38. [PMID: 12634353 PMCID: PMC150669 DOI: 10.1128/jvi.77.7.3929-3938.2003] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The dengue virus type 2 structures probably involved in human virulence were previously defined by sequencing the complete genome of both American and Southeast (SE) Asian genotype templates in patient serum (K. C. Leitmeyer et al., J. Virol. 73:4738-4747, 1999). We have now evaluated the effects of introducing a mutation in the envelope glycoprotein (E) gene and/or replacement of 5'- and 3'-nontranslated regions on dengue virus replication in human primary cell cultures. A series of chimeric infectious clones were generated containing different combinations of American and SE Asian genotype sequences. Some of the chimeric viruses had altered plaque morphology in mammalian cells; however, they replicated at similar rates in mosquito cells as measured by quantitative reverse transcription-PCR and plaque assay. Although susceptibility to virus infection varied from donor to donor in experiments using human macrophage and dendritic cells, we were able to measure consistent differences in viral RNA output per infected cell. Using this measurement, we demonstrated that the chimeric virus containing the E mutation had a lower virus output compared to the parental infectious clone. A larger reduction in virus output was observed for the triple mutant and the wild-type, American genotype virus from which chimeric inserts were derived. It appears that the three changes function synergistically, although the E mutation alone gives a lower output compared to the 5'- and 3'-terminal mutations. The data suggest that these changes may be responsible for decreased dengue virus replication in human target cells and for virulence characteristics during infection.
Collapse
Affiliation(s)
- Raymond Cologna
- Department of Virology and Immunology, Southwest Foundation for Biomedical Research, San Antonio, Texas 78227, USA
| | | |
Collapse
|
29
|
Wang WK, Chao DY, Kao CL, Wu HC, Liu YC, Li CM, Lin SC, Ho ST, Huang JH, King CC. High levels of plasma dengue viral load during defervescence in patients with dengue hemorrhagic fever: implications for pathogenesis. Virology 2003; 305:330-8. [PMID: 12573578 DOI: 10.1006/viro.2002.1704] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Studies of the pathogenesis of dengue hemorrhagic fever (DHF), a potentially life-threatening disease, have revealed the importance of initial high levels of virus replication. However, the possible involvement of virus during the transition from fever to defervescence, a critical stage in determining the severity of disease, has not been appreciated. Using quantitative reverse transcription-polymerase chain reaction, we examined the levels of plasma dengue viral load during both fever and defervescence periods in patients from a DEN-3 outbreak in southern Taiwan in 1998. Higher levels of plasma dengue viral RNA were found in DHF patients than in DF patients. During defervescence, while the level of plasma dengue viral RNA was undetectable in most DF patients, it remains high in all DHF patients. Using a modified immunoprecipitation assay, we demonstrated for the first time that the plasma dengue viruses persisting during defervescence were in the immune complexes for most DHF patients. These findings suggest that continued active viral replication or delay in the clearance of viremia contributes to the pathogenesis of DHF. Moreover, high levels of plasma dengue viral RNA during defervescence may serve as a disease marker for DHF.
Collapse
Affiliation(s)
- Wei-Kung Wang
- Institute of Microbiology, College of Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Within the flavivirus family, viruses that cause natural infections of the central nervous system (CNS) principally include members of the Japanese encephalitis virus (JEV) serogroup and the tick-borne encephalitis virus (TBEV) serocomplex. The pathogenesis of diseases involves complex interactions of viruses, which differ in neurovirulence potential, and a number of host factors, which govern susceptibility to infection and the capacity to mount effective antiviral immune responses both in the periphery and within the CNS. This chapter summarizes progress in the field of flavivirus neuropathogenesis. Mosquito-borne and tickborne viruses are considered together. Flavivirus neuropathogenesis involves both neuroinvasiveness (capacity to enter the CNS) and neurovirulence (replication within the CNS), both of which can be manipulated experimentally. Neuronal injury as a result of bystander effects may be a factor during flavivirus neuropathogenesis given that microglial activation and elaboration of inflammatory mediators, including IL-1β and TNF-α, occur in the CNS during these infections and may accompany the production of nitric oxide and peroxynitrite, which can cause neurotoxicity.
Collapse
Affiliation(s)
- Thomas J Chambers
- Department of Molecular Microbiology and Immunology, St. Louis University Health Sciences Center, School of Medicine, St. Louis, Missouri 63104, USA
| | | |
Collapse
|
31
|
Lin YW, Wang KJ, Lei HY, Lin YS, Yeh TM, Liu HS, Liu CC, Chen SH. Virus replication and cytokine production in dengue virus-infected human B lymphocytes. J Virol 2002; 76:12242-9. [PMID: 12414963 PMCID: PMC136880 DOI: 10.1128/jvi.76.23.12242-12249.2002] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dengue virus (DV) replication, antibody-enhanced viral infection, and cytokine responses of human primary B lymphocytes (cells) were characterized and compared with those of monocytes. The presence of a replication template (negative-strand RNA intermediate), viral antigens including core and nonstructural proteins, and increasing amounts of virus with time postinfection indicated that DV actively replicated in B cells. Virus infection also induced B cells to produce interleukin-6 and tumor necrosis factor alpha, which have been previously implicated in virus pathogenesis. In addition, a heterologous antibody was able to enhance both virus and cytokine production in B cells. Furthermore, the levels of virus replication, antibody-enhanced virus replication, and cytokine responses observed in B cells were not statistically different from those in monocytes. These results suggest that B cells may play an important role in DV pathogenesis.
Collapse
Affiliation(s)
- Yu-Wen Lin
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan 70101, Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Dengue virus (DV) replication, antibody-enhanced viral infection, and cytokine responses of human primary B lymphocytes (cells) were characterized and compared with those of monocytes. The presence of a replication template (negative-strand RNA intermediate), viral antigens including core and nonstructural proteins, and increasing amounts of virus with time postinfection indicated that DV actively replicated in B cells. Virus infection also induced B cells to produce interleukin-6 and tumor necrosis factor alpha, which have been previously implicated in virus pathogenesis. In addition, a heterologous antibody was able to enhance both virus and cytokine production in B cells. Furthermore, the levels of virus replication, antibody-enhanced virus replication, and cytokine responses observed in B cells were not statistically different from those in monocytes. These results suggest that B cells may play an important role in DV pathogenesis.
Collapse
|
33
|
Abstract
Severe dengue virus infections usually occur in individuals who have preexisting anti-dengue virus antibodies. Mast cells are known to play an important role in host defense against several pathogens, but their role in viral infection has not yet been elucidated. The effects of dengue virus infection on the production of chemokines by human mast cells were examined. Elevated levels of secreted RANTES, MIP-1alpha, and MIP-1beta, but not IL-8 or ENA-78, were observed following infection of KU812 or HMC-1 human mast cell-basophil lines. In some cases a >200-fold increase in RANTES production was observed. Cord blood-derived cultured human mast cells treated with dengue virus in the presence of subneutralizing concentrations of dengue virus-specific antibody also demonstrated significantly (P < 0.05) increased RANTES production, under conditions which did not induce significant degranulation. Chemokine responses were not observed when mast cells were treated with UV-inactivated dengue virus in the presence or absence of human dengue virus-specific antibody. Neither antibody-enhanced dengue virus infection of the highly permissive U937 monocytic cell line nor adenovirus infection of mast cells induced a RANTES, MIP-1alpha, or MIP-1beta response, demonstrating a selective mast cell response to dengue virus. These results suggest a role for mast cells in the initiation of chemokine-dependent host responses to dengue virus infection.
Collapse
Affiliation(s)
- Christine A King
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4H7, Canada
| | | | | |
Collapse
|
34
|
Germi R, Crance JM, Garin D, Guimet J, Lortat-Jacob H, Ruigrok RWH, Zarski JP, Drouet E. Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Virology 2002; 292:162-8. [PMID: 11878919 DOI: 10.1006/viro.2001.1232] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dengue virus type 2 and Yellow fever virus are arthropod-borne flaviviruses causing hemorrhagic fever in humans. Identification of virus receptors is important in understanding flavivirus pathogenesis. The aim of this work was to study the role of cellular heparan sulfate in the adsorption of infectious Yellow fever and Dengue type 2 viruses. Virus attachment was assessed by adsorbing virus to cells, washing unbound virus away, releasing cell-bound virus by freezing/thawing, and then titrating the released infectious virus. Treatment of cells by heparin-lyase, desulfation of cellular heparan sulfate, or treatment of the virus with heparin inhibited cell binding of both viruses. Heparin also inhibited Yellow fever virus infection by 97%. Using infectious virus, the present work shows the importance of heparan sulfate in binding and infection of these two flaviviruses.
Collapse
Affiliation(s)
- Raphaële Germi
- Laboratoire de Virologie Moléculaire et Structurale EA 2939, Université Joseph Fourier, Grenoble, La Tronche, 38700, France
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Diamond MS, Edgil D, Roberts TG, Lu B, Harris E. Infection of human cells by dengue virus is modulated by different cell types and viral strains. J Virol 2000; 74:7814-23. [PMID: 10933688 PMCID: PMC112311 DOI: 10.1128/jvi.74.17.7814-7823.2000] [Citation(s) in RCA: 208] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although prior studies have investigated cellular infection by dengue virus (DV), many have used highly passaged strains. We have reassessed cellular infection by DV type 2 (DV2) using prototype and low-passage isolates representing genotypes from different geographic areas. We observed marked variation in the susceptibility to infection among cell types by different DV2 strains. HepG2 hepatoma cells were susceptible to infection by all DV2 strains assayed. Although the prototype strain generated higher titers of secreted virus than the low-passage isolates, this difference did not correspond to positive- or negative-strand viral RNA levels and thus may reflect variation in efficiency among DV2 isolates to translate viral proteins or package and/or secrete virus. In contrast, human foreskin fibroblasts were susceptible to the prototype and low-passage Thai isolates but not to five Nicaraguan strains tested, as reflected by the absence of accumulation of negative-strand viral RNA, viral antigen, and infectious virus. A similar pattern was observed with the antibody-dependent pathway of infection. U937 and THP-1 myeloid cells and peripheral blood monocytes were infected in the presence of enhancing antibodies by the prototype strain but not by low-passage Nicaraguan isolates. Again, the barrier appeared to be prior to negative-strand accumulation. Thus, depending on the cell type and viral isolate, blocks that limit the production of infectious virus in vitro may occur at distinct steps in the pathway of cellular infection.
Collapse
Affiliation(s)
- M S Diamond
- Division of Infectious Diseases, School of Public Health, University of California, Berkeley, California 94720, USA
| | | | | | | | | |
Collapse
|
36
|
Diamond MS, Roberts TG, Edgil D, Lu B, Ernst J, Harris E. Modulation of Dengue virus infection in human cells by alpha, beta, and gamma interferons. J Virol 2000; 74:4957-66. [PMID: 10799569 PMCID: PMC110847 DOI: 10.1128/jvi.74.11.4957-4966.2000] [Citation(s) in RCA: 274] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A role for interferon (IFN) in modulating infection by dengue virus (DV) has been suggested by studies in DV-infected patients and IFN receptor-deficient mice. To address how IFN modulates DV type 2 infection, we have assayed IFN-alpha, -beta, and -gamma for the ability to enhance or diminish antibody-independent and antibody-dependent cell infection using a competitive, asymmetric reverse transcriptase-mediated PCR (RT-PCR) assay that quantitates positive and negative strands of viral RNA, a flow cytometric assay that measures viral antigen, and a plaque assay that analyzes virion production. Our data suggest that IFN-alpha and -beta protect cells against DV infection in vitro. Treatment of hepatoma cells with IFN-alpha or -beta decreases viral RNA levels greater than 1, 000-fold, the percentage of cells infected 90 to 95%, and the amount of infectious virus secreted 150- to 100,000-fold. These results have been reproduced with several cell types and viral strains, including low-passage isolates. In contrast, IFN-gamma has a more variable effect depending on the cell type and pathway of infection. Quantitative RT-PCR experiments indicate that IFN inhibits DV infection by preventing the accumulation of negative-strand viral RNA.
Collapse
Affiliation(s)
- M S Diamond
- Division of Infectious Diseases, School of Public Health, University of California, Berkeley, California 94720, USA
| | | | | | | | | | | |
Collapse
|
37
|
Lin YL, Liao CL, Chen LK, Yeh CT, Liu CI, Ma SH, Huang YY, Huang YL, Kao CL, King CC. Study of Dengue virus infection in SCID mice engrafted with human K562 cells. J Virol 1998; 72:9729-37. [PMID: 9811707 PMCID: PMC110483 DOI: 10.1128/jvi.72.12.9729-9737.1998] [Citation(s) in RCA: 146] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/1998] [Accepted: 09/09/1998] [Indexed: 11/20/2022] Open
Abstract
Here we report that severe combined immunodeficient (SCID) mice engrafted with human K562 cells (K562-SCID mice) can be used as an animal model to study dengue virus (DEN) infection. After intratumor injection into K562 cell masses of PL046, a Taiwanese DEN-2 human isolate, the K562-SCID mice showed neurological signs of paralysis and died at approximately 2 weeks postinfection. In addition to being detected in the tumor masses, high virus titers were detected in the peripheral blood and the brain tissues, indicating that DEN had replicated in the infected K562-SCID mice. In contrast, the SCID mice were resistant to DEN infection and the mock-infected K562-SCID mice survived for over 3 months. These data illustrate that DEN infection contributed directly to the deaths of the infected K562-SCID mice. Other serotypes of DEN were also used to infect the K562-SCID mice, and the mortality rates of the infected mice varied with different challenge strains, suggesting the existence of diverse degrees of virulence among DENs. To determine whether a neutralizing antibody against DEN in vitro was also protective in vivo, the K562-SCID mice were challenged with DEN-2 and received antibody administration at the same time or 1 day earlier. Our results revealed that the antibody-treated mice exhibited a reduction in mortality and a delay of paralysis onset after DEN infection. In contrast to K562-SCID, the persistently DEN-infected K562 cells generated in vitro invariably failed to be implanted in the mice. It seems that in the early stage of implantation, a gamma interferon activated, nitric oxide-mediated anti-DEN effect might play a role in the innate immunity against DEN-infected cells. The system described herein offers an opportunity to explore DEN replication in vivo and to test various antiviral protocols in infected hosts.
Collapse
Affiliation(s)
- Y L Lin
- Institute of Biomedical Sciences, Academia Sinica, National Taiwan University, Taipei, Taiwan, Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Dengue fever, a very old disease, has reemerged in the past 20 years with an expanded geographic distribution of both the viruses and the mosquito vectors, increased epidemic activity, the development of hyperendemicity (the cocirculation of multiple serotypes), and the emergence of dengue hemorrhagic fever in new geographic regions. In 1998 this mosquito-borne disease is the most important tropical infectious disease after malaria, with an estimated 100 million cases of dengue fever, 500,000 cases of dengue hemorrhagic fever, and 25,000 deaths annually. The reasons for this resurgence and emergence of dengue hemorrhagic fever in the waning years of the 20th century are complex and not fully understood, but demographic, societal, and public health infrastructure changes in the past 30 years have contributed greatly. This paper reviews the changing epidemiology of dengue and dengue hemorrhagic fever by geographic region, the natural history and transmission cycles, clinical diagnosis of both dengue fever and dengue hemorrhagic fever, serologic and virologic laboratory diagnoses, pathogenesis, surveillance, prevention, and control. A major challenge for public health officials in all tropical areas of the world is to develop and implement sustainable prevention and control programs that will reverse the trend of emergent dengue hemorrhagic fever.
Collapse
Affiliation(s)
- D J Gubler
- Division of Vector-Borne Infectious Diseases, Centers for Disease Control and Prevention, Fort Collins, Colorado 80522, USA.
| |
Collapse
|
39
|
Jänisch T, Preiser W, Berger A, Niedrig M, Mikulicz U, Thoma B, Doerr HW. Emerging viral pathogens in long-term expatriates (II): Dengue virus. Trop Med Int Health 1997; 2:934-40. [PMID: 9357483 DOI: 10.1046/j.1365-3156.1997.d01-149.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dengue virus infections have been well known for many years; still dengue virus is regarded as an 'emerging' pathogen, as the disease profile is changing. Its geographical range and overall incidence, and the incidence of the associated complications, dengue haemorrhagic fever (DHF) and dengue shock syndrome (DSS), are on the increase. Modern-day travel and increasing urbanization seem to be the main contributing factors. In order to estimate the risk of infection during long-term stays in dengue-endemic countries, we tested sera obtained from 323 development aid workers and their family members who had spent on average 9.8 years in dengue-endemic regions for the presence of dengue virus antibodies. Dengue virus antibody screening was done by a commercially available immunofluorescence test (IF). Reactive samples were re-tested by an in-house IF and also tested for cross-reactivity to yellow fever virus using yellow fever IF and neutralization test (NT). Evaluation of the results revealed that the screening test has a specificity of at least 63.2%. In 12 of 19 initially positive cases crossreacting antibodies against yellow fever virus could be ruled out. Three cases remained indeterminable, whereas four of the reactive and 10 (out of 12) of the borderline reactive cases showed crossreactivity with yellow fever virus, probably due to previous vaccination. We found seroprevalence rates of 4.3% with no significant differences related to gender or area of upbringing. Seroprevalence rates were evaluated according to region of suspected or confirmed infection. In two cases the dengue infection had taken a classical clinical course; in another three cases an extraordinary febrile illness was reported in the history. None of the other seropositive individuals had a history of an illness possibly attributable to dengue virus infection. Our results show that there definitely is a risk for long-term expatriates to acquire (mostly non- or oligo-symptomatic) dengue infection, which might be important especially in the light of the supposed aetiology of DHF or DSS as a secondary infection with another dengue virus serotype.
Collapse
Affiliation(s)
- T Jänisch
- Institut für Medizinische Virologie, Klinikum der J. W. Goethe-Universität, Frankfurt am Main, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Anderson R, Wang S, Osiowy C, Issekutz AC. Activation of endothelial cells via antibody-enhanced dengue virus infection of peripheral blood monocytes. J Virol 1997; 71:4226-32. [PMID: 9151809 PMCID: PMC191637 DOI: 10.1128/jvi.71.6.4226-4232.1997] [Citation(s) in RCA: 149] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Although endothelial cells have been speculated to be a target in the pathogenesis of dengue hemorrhagic fever (DHF), there has been little evidence linking dengue virus infection to any alteration in endothelial cell function. In this study, we show that human umbilical vein endothelial cells become activated when exposed to culture fluids from dengue virus-infected peripheral blood monocytes. Maximum activation was achieved with culture fluids from monocytes in which virus infection was enhanced by the addition of dengue virus-immune serum, thus correlating with epidemiological evidence that prior immunity to dengue virus is a major risk factor for DHF. Activation was strongest for endothelial cell expression of VCAM-1 and ICAM-1. In contrast, activation of endothelial cell E-selectin expression appeared to be more transient, as indicated by its detection at 3 h, but not at 16 h, of treatment. Treatment of monocyte culture fluids with anti-tumor necrosis factor alpha (TNF-alpha) antibody largely abolished the activation effect (as measured by endothelial cell expression of ICAM-1), whereas treatment with IL-1beta receptor antagonist had a much smaller inhibitory effect on activation. Endothelial cells inoculated directly with dengue virus or with virus-antibody combinations were poorly infectable (compared to Vero cells or peripheral blood monocytes), and virus-inoculated endothelial cells showed no increased expression of VCAM-1, ICAM-1, or E-selectin. Taken together, the results strongly indicate that dengue virus can modulate endothelial cell function by an indirect route, in which a key intermediary is TNF-alpha released from virus-infected monocytes.
Collapse
Affiliation(s)
- R Anderson
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada.
| | | | | | | |
Collapse
|
41
|
Abstract
Many virus infections in man and other species are accompanied by immunosuppression. This is clearly important in terms of the susceptibility of the host to secondary infections. The immunosuppression may also aid and abet the growth and persistence of viruses. An unresolved issue is the extent to which the extent of this immunosuppression is determined by the virulence of the infecting virus or resistance factors in the host, and particularly by factors that are genetically determined. The mechanisms of viral immunosuppression are indirect and direct. Indirect mechanisms such as interferon production and suppressor cells induced by infection undoubtedly contribute to viral immunosuppression in experimental models of virus infection. In man the direct inactivation of immunologically responsive lymphocytes seems to be the most important mechanism. Moreover, the persistence of viruses in human lymphocytes is being increasingly recognized.
Collapse
|
42
|
He RT, Innis BL, Nisalak A, Usawattanakul W, Wang S, Kalayanarooj S, Anderson R. Antibodies that block virus attachment to Vero cells are a major component of the human neutralizing antibody response against dengue virus type 2. J Med Virol 1995; 45:451-61. [PMID: 7666046 DOI: 10.1002/jmv.1890450417] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Epidemiological data strongly implicate a role for the host humoral immune response in both protection against and exacerbation of dengue virus-caused disease. In an effort to characterize elements of the normal human immune response against dengue virus we have addressed the issue of antibody-mediated neutralization of dengue virus. We show here the ability of both mouse monoclonal antibody 3H5 and human anti-dengue neutralizing sera to block binding of dengue-2 virus to monkey kidney (Vero) cells. Since Vero cells possess virus receptors but not Fc receptors we conclude that the major effect of host neutralizing antibodies is to block virus attachment to Vero cell dengue virus receptors. Analysis of 61 patient antisera yielded good correlation (Pearson's coefficient = 0.90; P < 0.001) between neutralizing activity and ability to block virus-cell attachment suggesting that antibody-mediated neutralization of dengue virus occurs primarily extracellularly and less by a postattachment mechanism as has been described for certain other viruses.
Collapse
Affiliation(s)
- R T He
- Department of Microbiology and Infectious Diseases, University of Calgary, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Ochiai H, Kurokawa M, Matsui S, Yamamoto T, Kuroki Y, Kishimoto C, Shiraki K. Infection enhancement of influenza A NWS virus in primary murine macrophages by anti-hemagglutinin monoclonal antibody. J Med Virol 1992; 36:217-21. [PMID: 1564451 DOI: 10.1002/jmv.1890360312] [Citation(s) in RCA: 51] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Antibody-dependent enhancement (ADE) of influenza A NWS virus infection was investigated in primary murine macrophages (M phi) using anti-hemagglutinin(HA) monoclonal antibody (mAB). Contrary to previous reports of abortive influenza virus infection in primary M phi, this study demonstrated that the NWS virus replicated productively in both resident peritoneal M phi and thioglycolate-elicited peritoneal M phi providing cleavage of the HA was achieved by trypsin; 5 micrograms/ml of trypsin was the optimum concentration for the induction of infectivity. Under multiple-cycle growth conditions in the presence of mAB at various concentrations in trypsin-containing media, ADE was demonstrated in both M phi in the presence of subneutralizing concentrations of mAB. Flow cytometric analysis showed that the mechanism of virus entry into M phi could be through HA to specific virus receptors, or HA plus antibody to Fc receptors. These results indicate that ADE of the NWS virus infection actually occurs on Fc receptor-bearing primary murine M phi depending on the concentration of antibody in the presence of the appropriate protease for cleavage of viral HA.
Collapse
Affiliation(s)
- H Ochiai
- Department of Virology, Toyama Medical and Pharmaceutical University, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Abstract
Titration experiments in swine macrophages have shown that African swine fever virus infectivity was not enhanced in the presence of antiviral antibodies. The early viral protein synthesis and the viral DNA replication in swine macrophages infected with virus-antibody complexes were inhibited in the presence of high doses of uv-inactivated virus, which saturated specific virus receptors, but not when Fc receptors were saturated with antibodies. These results indicate that African swine fever virus does not infect swine macrophages through Fc receptors and that the normal entry pathway through virus receptors is not bypassed by the virus-antibody complexes.
Collapse
Affiliation(s)
- A Alcamí
- Centro de Biología Molecular (CSIC-UAM), Facultad de Ciencias, Universidad Autónoma, Madrid, Spain
| | | |
Collapse
|
46
|
Hohdatsu T, Nakamura M, Ishizuka Y, Yamada H, Koyama H. A study on the mechanism of antibody-dependent enhancement of feline infectious peritonitis virus infection in feline macrophages by monoclonal antibodies. Arch Virol 1991; 120:207-17. [PMID: 1659798 PMCID: PMC7087175 DOI: 10.1007/bf01310476] [Citation(s) in RCA: 61] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Enhancement of feline infectious peritonitis virus (FIPV) infection of feline macrophages was studied using monoclonal antibodies (MAbs) to the FIPV strain 79-1146. Adherent cells recovered from the feline lung and peritoneal cavity phagocytosed fixed red blood cells, and formed Fc-mediated rosettes. Enhancement of virus infection by MAb was investigated by inoculating alveolar macrophages with a mixtures of viral suspension and MAb, and examining the cells for intracellular viral antigen by the immunofluorescence assay and the amount of infectious virus in the supernatant fluid after incubation. The replication of FIPV in macrophages was enhanced by non-neutralizing MAbs recognizing peplomer protein (S) and transmembrane protein (M) of the virus. Even among the MAbs having the ability to neutralize FIPV strain 79-1146, some reversely enhanced virus infection when they were diluted. The enhancement was suppressed by pretreatment of the MAb with protein A. The enhancement was reduced by the use of F(ab')2 fragment of MAb. These results demonstrated antibody-dependent enhancement (ADE) of FIPV infection in macrophage. The replication of FIPV 79-1146 strain in macrophages from FIPV antibody-positive cats was more enhanced than in those from antibody-negative cats.
Collapse
Affiliation(s)
- T Hohdatsu
- Department of Veterinary Infectious Diseases, School of Veterinary Medicine and Animal Sciences, Kitasato University, Aomori-Ken, Japan
| | | | | | | | | |
Collapse
|
47
|
Abstract
Although CD4 is a major receptor for human immunodeficiency virus (HIV) infection of cells, studied by ourselves and others clearly show that the Fc receptor (FcR) also plays a role in infection, perhaps in conjunction with other surface receptors. IgG antibodies to HIV-1 will enhance infectivity in cells (such as monocyte-macrophages) that have surface Fc receptors; F(ab')2 fragments of antibodies did not enhance, and blocking of FcR inhibited enhancement. The high-affinity FcR for IgG (Fc gamma RI) appeared to be functional. Sera from HIV-1-infected patients had neutralizing activity at high concentrations, but enhanced infection at low concentrations (i.e., high dilutions). Our studies show that the CD4 receptor is required for antibody-mediated enhancement of infection, as enhancement can be blocked by recombinant soluble CD4 and by Leu3 antibody. Although enhancement can be demonstrated in vitro, the in vivo importance of enhancing antibodies remains to be defined in HIV-1 infection.
Collapse
Affiliation(s)
- A Takeda
- Department of Medicine, University of Massachusetts Medical School, Worcester 01655
| | | |
Collapse
|
48
|
Ochiai H, Kurokawa M, Kuroki Y, Niwayama S. Infection enhancement of influenza A H1 subtype viruses in macrophage-like P388D1 cells by cross-reactive antibodies. J Med Virol 1990; 30:258-65. [PMID: 2370521 DOI: 10.1002/jmv.1890300406] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The contribution of cross-reactive hemagglutination inhibition (HI) antibodies to infection enhancement of influenza A H1 subtype NWS virus and two antigenic drift strains was investigated in a macrophage-like cell line P388D1. When P388D1 cells, previously treated with neuraminidase (NA) to remove the viral receptors, were infected with NWS virus exposed to rabbit antiviral immunoglobulin (IgG) showing various levels of cross-HI titers, virus yields were enhanced in the presence of a subneutralizing antibody, depending on their cross-HI titers. By flow cytometric analysis using a fluorescein isothiocyanate (FITC)-labeled NWS virus, the efficiency of attachment of virus-rabbit IgG complexes to Fc receptors on NA-treated cells showed close correlation with its cross-HI titer. These data suggest that cross-reactive HI antibodies could contribute to infection enhancement through the formation of potent infectious immune complexes with drift strains to mediate virus infection via Fc receptor uptake. Two monoclonal antibodies (mAB) in mouse IgG subclasses IgG1 and IgG2a showing strain-specific or cross-reactive HI activity were tested for their infection enhancement characteristics. A strain-specific mAB enhanced infection of homologous NWS virus, but not that of two other drift strains in either antibody dilution. In contrast, a cross-reactive mAB caused infection enhancement of all three virus strains in the presence of the subneutralizing antibody. This indicates that cross-reactivity, but not the IgG subclass, acts as an enhancing factor to this phenomenon. The antibody, with the same specificity as cross-reactive mAB, was detected semiquantitatively by competitive enzyme-linked immunosorbent assay (ELISA) with results almost consistent with cross-HI titers of polyclonal rabbit antiviral IgGs. These data suggest that the antibody detected by this assay might be one of the potent antibodies governing cross-HI activity as a whole antibody and causing infection enhancement of drift strains.
Collapse
Affiliation(s)
- H Ochiai
- Department of Virology, Toyama Medical and Pharmaceutical University, Japan
| | | | | | | |
Collapse
|
49
|
Kurane I, Kontny U, Janus J, Ennis FA. Dengue-2 virus infection of human mononuclear cell lines and establishment of persistent infections. Arch Virol 1990; 110:91-101. [PMID: 2178591 DOI: 10.1007/bf01310705] [Citation(s) in RCA: 58] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Twenty three human mononuclear cell lines including ten myelomonocytic cell lines, eight B cell lines and five T cell lines, were examined to determine whether they could be infected with dengue-2 virus. All the cell lines were infected with dengue-2 virus as determined by immunofluorescent staining and by virus titration of culture supernatant fluids. K562, Jiyoye and Jurkat, respectively, showed the highest percentage of infected cells of these myelomonocytic, B and T cell lines. Antibody to dengue-2 virus at subneutralizing concentrations augmented dengue-2 virus infection of myelomonocytic cell lines, but not of B cell lines or of T cell lines. Persistent dengue-2 virus infection was established using a myelomonocytic cell line (K562), a B cell line (Raji), and a T cell line (HSB-2). These cell lines maintained a high percentage (more than 70%) of dengue-2 virus antigen-positive cells for at least 25 weeks. Very low titers of infectious dengue-2 virus were detected in the culture supernatant fluids of the persistently infected cells. Dengue-2 virus antigen-positive Raji cell clones were established from persistently-infected Raji cells using limiting dilutions and all of the cells in these clones were dengue-2 virus antigen-positive. These findings demonstrate that a variety of human mononuclear cell lines can be infected with dengue-2 virus and may be useful as models for the analysis of dengue virus-human cell interactions in dengue virus infections.
Collapse
Affiliation(s)
- I Kurane
- Department of Medicine, University of Massachusetts Medical Center, Worcester
| | | | | | | |
Collapse
|
50
|
|