401
|
From research to phase III: Preclinical, industrial and clinical development of the Sanofi Pasteur tetravalent dengue vaccine. Vaccine 2011; 29:7229-41. [DOI: 10.1016/j.vaccine.2011.06.094] [Citation(s) in RCA: 231] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/16/2011] [Accepted: 06/24/2011] [Indexed: 02/06/2023]
|
402
|
Cross-reactivity and expansion of dengue-specific T cells during acute primary and secondary infections in humans. Sci Rep 2011; 1:51. [PMID: 22355570 PMCID: PMC3216538 DOI: 10.1038/srep00051] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 07/18/2011] [Indexed: 12/17/2022] Open
Abstract
Serotype-cross-reactive memory T cells responding to secondary dengue virus (DENV) infection are thought to contribute to disease. However, epitope-specific T cell responses have not been thoroughly compared between subjects with primary versus secondary DENV infection. We studied CD8(+) T cells specific for the HLA-A*1101-restricted NS3(133) epitope in a cohort of A11(+) DENV-infected patients throughout acute illness and convalescence. We compared the expansion, serotype-cross-reactivity, and activation of these cells in PBMC from patients experiencing primary or secondary infection and mild or severe disease by flow cytometry. Our results show expansion and activation of DENV-specific CD8(+) T cells during acute infection, which are predominantly serotype-cross-reactive regardless of DENV infection history. These data confirm marked T cell activation and serotype-cross-reactivity during the febrile phase of dengue; however, A11-NS3(133)-specific responses did not correlate with prior antigenic exposure or current disease severity.
Collapse
|
403
|
Abstract
Dengue is an important cause of childhood and adult morbidity in Asian and Latin American countries and its geographic footprint is growing. The clinical manifestations of dengue are the expression of a constellation of host and viral factors, some acquired, others intrinsic to the individual. The virulence of the virus plus the flavivirus infection history, age, gender and genotype of the host all appear to help shape the severity of infection. Similarly, the characteristics of the innate and acquired host immune response subsequent to infection are also likely determinants of outcome. This review summarises recent developments in the understanding of dengue pathogenesis and their relevance to dengue vaccine development.
Collapse
|
404
|
Rothman AL. Immunity to dengue virus: a tale of original antigenic sin and tropical cytokine storms. Nat Rev Immunol 2011; 11:532-43. [PMID: 21760609 DOI: 10.1038/nri3014] [Citation(s) in RCA: 562] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dengue is a mosquito-borne viral disease of expanding geographical range and incidence. The existence of four viral serotypes and the association of prior dengue virus infection with an increased risk for more severe disease have presented significant obstacles to vaccine development. An increased understanding of the adaptive immune response to natural dengue virus infection and candidate dengue vaccines has helped to define the specific antibody and T cell responses that are associated with either protective or pathological immunity during dengue infection. Further characterization of immunological correlates of disease outcome and the validation of these findings in vaccine trials will be invaluable for developing effective dengue vaccines.
Collapse
Affiliation(s)
- Alan L Rothman
- Institute for Immunology and Informatics and Department of Cell and Molecular Biology, University of Rhode Island, Providence, Rhode Island 02903, USA.
| |
Collapse
|
405
|
Terajima M, Ennis FA. T cells and pathogenesis of hantavirus cardiopulmonary syndrome and hemorrhagic fever with renal syndrome. Viruses 2011; 3:1059-73. [PMID: 21994770 PMCID: PMC3185782 DOI: 10.3390/v3071059] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 06/30/2011] [Accepted: 07/01/2011] [Indexed: 12/17/2022] Open
Abstract
We previously hypothesized that increased capillary permeability observed in both hantavirus cardiopulmonary syndrome (HCPS) and hemorrhagic fever with renal syndrome (HFRS) may be caused by hantavirus-specific cytotoxic T cells attacking endothelial cells presenting viral antigens on their surface based on clinical observations and in vitro experiments. In HCPS, hantavirus-specific T cell responses positively correlated with disease severity. In HFRS, in one report, contrary to HCPS, T cell responses negatively correlated with disease severity, but in another report the number of regulatory T cells, which are thought to suppress T cell responses, negatively correlated with disease severity. In rat experiments, in which hantavirus causes persistent infection, depletion of regulatory T cells helped infected rats clear virus without inducing immunopathology. These seemingly contradictory findings may suggest delicate balance in T cell responses between protection and immunopathogenesis. Both too strong and too weak T cell responses may lead to severe disease. It is important to clarify the role of T cells in these diseases for better treatment (whether to suppress T cell functions) and protection (vaccine design) which may need to take into account viral factors and the influence of HLA on T cell responses.
Collapse
Affiliation(s)
- Masanori Terajima
- Center for Infectious Disease and Vaccine Research, Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | |
Collapse
|
406
|
Dejnirattisai W, Webb AI, Chan V, Jumnainsong A, Davidson A, Mongkolsapaya J, Screaton G. Lectin switching during dengue virus infection. J Infect Dis 2011; 203:1775-83. [PMID: 21606536 PMCID: PMC3100511 DOI: 10.1093/infdis/jir173] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/24/2011] [Indexed: 12/29/2022] Open
Abstract
Dengue virus receptors are relatively poorly characterized, but there has been recent interest in 2 C-type lectin molecules, dendritic cell-specific intercellular adhesion molecule 3 (ICAM-3)-grabbing nonintegrin (DC-SIGN) and its close homologue liver/lymph node-specific ICAM-3-grabbing integrin (L-SIGN), which can both bind dengue and promote infection. In this report we have studied the interaction of dengue viruses produced in insect cells, tumor cell lines, and primary human dendritic cells (DCs) with DC-SIGN and L-SIGN. Virus produced in primary DCs is unable to interact with DC-SIGN but remains infectious for L-SIGN-expressing cells. Skin-resident DCs may thus be a site of initial infection by insect-produced virus, but DCs will likely not participate in large-scale virus replication during dengue infection. These results reveal that differential glycosylation of dengue virus envelope protein is highly dependent on cell state and suggest that studies of virus tropism using virus prepared in insect cells or tumor cell lines should be interpreted with caution.
Collapse
Affiliation(s)
- Wanwisa Dejnirattisai
- Department of Medicine, Faculty of Medicine, Imperial College London, United Kingdom
| | - Andrew I. Webb
- Department of Medicine, Faculty of Medicine, Imperial College London, United Kingdom
| | - Vera Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong
| | - Amonrat Jumnainsong
- Department of Medicine, Faculty of Medicine, Imperial College London, United Kingdom
| | - Andrew Davidson
- Department of Cellular and Molecular Medicine, University of Bristol, United Kingdom
| | - Juthathip Mongkolsapaya
- Department of Medicine, Faculty of Medicine, Imperial College London, United Kingdom
- Dengue Hemorrhagic Fever Research Unit Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Gavin Screaton
- Department of Medicine, Faculty of Medicine, Imperial College London, United Kingdom
| |
Collapse
|
407
|
Abstract
Feline infectious peritonitis (FIP) is a fatal, immune-augmented, and progressive viral disease of cats associated with feline coronavirus (FCoV). Viral genetic determinants specifically associated with FIPV pathogenesis have not yet been discovered. Viral gene signatures in the spike, non-structural protein 3c, and membrane of the coronavirus genome have been shown to often correlate with disease manifestation. An “in vivo mutation transition hypothesis” is widely accepted and postulates that de novo virus mutation occurs in vivo giving rise to virulence. The existence of “distinct circulating avirulent and virulent strains” is an alternative hypothesis of viral pathogenesis. It may be possible that viral dynamics from both hypotheses are at play in the occurrence of FIP. Epidemiologic data suggests that the genetic background of the cat contributes to the manifestation of FIP. Further studies exploring both viral and host genetic determinants of disease in FIP offer specific opportunities for the management of this disease.
Collapse
Affiliation(s)
- Meredith A Brown
- New Mexico State University, Department of Biology, Las Cruces, NM 88003-8001, USA.
| |
Collapse
|
408
|
Abstract
Dengue virus (DENV) is a mosquito-borne member of the Flavivirus genus and includes four serotypes (DENV-1, DENV-2, DENV-3, and DENV-4), each of which is capable of causing dengue fever and dengue hemorrhagic fever/dengue shock syndrome. Serious disease can be seen during primary infection but is more frequent following second infection with a serotype different from that of a previous infection. Infection with wild-type DENV induces high-titered neutralizing antibody that can provide long-term immunity to the homotypic virus and can provide short-term immunity (only several months duration) to a heterotypic DENV. The high level of virus replication seen during both secondary infection with a heterotypic virus and during primary DENV infection in late infancy is a direct consequence of antibody-dependent enhancement of replication. This enhanced virus replication is mediated primarily by preexisting, nonneutralizing, or subneutralizing antibodies to the virion surface antigens that enhance access of the virion-antibody complex to FcγR-bearing cells. Vaccines will need to provide long-term protection against each of the four DENV serotypes by inducing neutralizing antibodies, and live, attenuated and various nonliving virus vaccines are in development.
Collapse
Affiliation(s)
- Brian R Murphy
- Laboratory of Infectious Diseases, National Institutes of Allergy and Infectious Diseases, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
409
|
Guzman MG, Halstead SB, Artsob H, Buchy P, Farrar J, Gubler DJ, Hunsperger E, Kroeger A, Margolis HS, Martínez E, Nathan MB, Pelegrino JL, Simmons C, Yoksan S, Peeling RW. Dengue: a continuing global threat. Nat Rev Microbiol 2011; 8:S7-16. [PMID: 21079655 DOI: 10.1038/nrmicro2460] [Citation(s) in RCA: 1264] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dengue fever and dengue haemorrhagic fever are important arthropod-borne viral diseases. Each year, there are ∼50 million dengue infections and ∼500,000 individuals are hospitalized with dengue haemorrhagic fever, mainly in Southeast Asia, the Pacific and the Americas. Illness is produced by any of the four dengue virus serotypes. A global strategy aimed at increasing the capacity for surveillance and outbreak response, changing behaviours and reducing the disease burden using integrated vector management in conjunction with early and accurate diagnosis has been advocated. Antiviral drugs and vaccines that are currently under development could also make an important contribution to dengue control in the future.
Collapse
Affiliation(s)
- Maria G Guzman
- Instituto de Medicina Tropical, 'Pedro Kouri', PO Box 601, Marianao 13, Ciucad de la Habana, Cuba
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
410
|
Coller BAG, Clements DE. Dengue vaccines: progress and challenges. Curr Opin Immunol 2011; 23:391-8. [PMID: 21514129 DOI: 10.1016/j.coi.2011.03.005] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 10/18/2022]
Abstract
With several dengue vaccine candidates progressing through clinical trials, several options for controlling this disease appear feasible. This would represent a major achievement and reflect decades of research and development activities. The challenges associated with the limited understanding of protective responses and those factors which determine disease severity remain, but with prospective studies ongoing in various dengue endemic areas and the initiation of dengue vaccine efficacy trials, immune responses are being evaluated in the context of protection and severe disease and these studies are highly likely to provide additional insights.
Collapse
Affiliation(s)
- Beth-Ann G Coller
- Vaccines Research, WP17-2131, Merck and Company, 770 Sumneytown Pike, P.O. Box 4, West Point, PA 19486, United States.
| | | |
Collapse
|
411
|
Gunther VJ, Putnak R, Eckels KH, Mammen MP, Scherer JM, Lyons A, Sztein MB, Sun W. A human challenge model for dengue infection reveals a possible protective role for sustained interferon gamma levels during the acute phase of illness. Vaccine 2011; 29:3895-904. [PMID: 21443963 DOI: 10.1016/j.vaccine.2011.03.038] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 03/11/2011] [Indexed: 11/17/2022]
Abstract
Dengue has recently been defined by the World Health Organization as a major international public health concern. Although several vaccine candidates are in various stages of development, there is no licensed vaccine available to assist in controlling the further spread of this mosquito borne disease. The need for a reliable animal model for dengue disease increases the risk to vaccine developers as they move their vaccine candidates into large-scale phase III testing. In this paper we describe the cellular immune responses observed in a human challenge model for dengue infection; a model that has the potential to provide efficacy data for potential vaccine candidates in a controlled setting. Serum levels of sIL-2Rα and sTNF-RII were increased in volunteers who developed illness. Supernatants from in vitro stimulated PBMC were tested for cytokines associated with a T(H)1 or T(H)2 T-cell response (IL-2, TNF-α, IFN-γ, IL-4, IL-10, IL-5) and only IFN-γ was associated with protection against fever and/or viremia. Interestingly, IFN-γ levels drop to 0 pg/mL for volunteers who develop illness after challenge suggesting that some mechanism of immunosuppression may play a role in dengue illness. The human challenge model provides an opportunity to test potential vaccine candidates for efficacy prior to large-scale phase III testing, and hints at a possible mechanism for immune suppression by dengue.
Collapse
Affiliation(s)
- V J Gunther
- Division of Viral Diseases, Walter Reed Army Institute of Research (WRAIR), Silver Spring, MD 20910, United States.
| | | | | | | | | | | | | | | |
Collapse
|
412
|
Dengue virus type 3 isolated from a fatal case with visceral complications induces enhanced proinflammatory responses and apoptosis of human dendritic cells. J Virol 2011; 85:5374-83. [PMID: 21450836 DOI: 10.1128/jvi.01915-10] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A recent (2007 to 2009) dengue outbreak caused by dengue virus (DENV) in Paraguay presented unusual severe clinical outcomes associated with 50% mortality rates. Although it has been reported that inflammatory responses influence the severity of dengue virus infection (T. Pang, M. J. Cardosa, and M. G. Guzman, Immunol. Cell Biol. 85:43-45, 2007), there remains a paucity of information on virus-innate immunity interactions influencing clinical outcome. Using human dendritic cells from a major innate immune cell population as an in vitro model, we have investigated signature cytokine responses as well as infectivity-replicative profiles of DENV clinical isolates from either a nonfatal case of classical dengue fever (strain DENV3/290; isolated in Brazil in 2002) or a fatal case of dengue fever with visceral complications isolated in Paraguay in 2007 (strain DENV3/5532). Strain DENV3/5532 was found to display significantly higher replicative ability than DENV3/290 in monocyte-derived dendritic cells (mdDCs). In addition, compared to DENV3/290 results, mdDCs exposed to DENV3/5532 showed increased production of proinflammatory cytokines associated with higher rates of programmed cell death, as shown by annexin V staining. The observed phenotype was due to viral replication, and tumor necrosis factor alpha (TNF-α) appears to exert a protective effect on virus-induced mdDC apoptosis. These results suggest that the DENV3/5532 strain isolated from the fatal case replicates within human dendritic cells, modulating cell survival and synthesis of inflammatory mediators.
Collapse
|
413
|
Duyen HTL, Ngoc TV, Ha DT, Hang VTT, Kieu NTT, Young PR, Farrar JJ, Simmons CP, Wolbers M, Wills BA. Kinetics of plasma viremia and soluble nonstructural protein 1 concentrations in dengue: differential effects according to serotype and immune status. J Infect Dis 2011; 203:1292-300. [PMID: 21335562 PMCID: PMC3069728 DOI: 10.1093/infdis/jir014] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We describe the magnitude and kinetics of plasma viremia and nonstructural protein 1 (sNS1) levels in sequential samples from 167 children with acute dengue, enrolled early in a community study in Vietnam. All children recovered fully, and only 5 required hospitalization. Among those with dengue virus type 1 (DENV-1), plasma viremia was significantly greater in primary (49) than secondary (44) infections and took longer to resolve. In primary DENV-2 and 3 infections, viremia was significantly lower than among primary DENV-1 infections. Concentrations of sNS1 were significantly higher for DENV-1 than for DENV-2 after adjusting for viremia, with marked differences in the kinetic profiles between primary and secondary infections. Secondary infection and higher viremia were independent predictors of more severe thrombocytopenia, and higher viremia was associated with a small increase in hemoconcentration. Our findings identify clear serotype and immune-status related effects on the dynamics of dengue viremia and sNS1 responses, together with associations with important clinical parameters.
Collapse
Affiliation(s)
- Huynh T L Duyen
- Oxford University Clinical Research Unit, Churchill Hospital, Oxford, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
414
|
Sun P, Beckett C, Danko J, Burgess T, Liang Z, Kochel T, Porter K. A dendritic cell-based assay for measuring memory T cells specific to dengue envelope proteins in human peripheral blood. J Virol Methods 2011; 173:175-81. [PMID: 21315764 DOI: 10.1016/j.jviromet.2011.01.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Revised: 01/20/2011] [Accepted: 01/31/2011] [Indexed: 11/18/2022]
Abstract
Dengue envelope (E) protein is a dominant immune inducer and E protein-based vaccines elicited partial to complete protection in non-human primates. To study the immunogenicity of these vaccines in humans, an enzyme linked immunospot (ELISPOT) assay for measuring interferon gamma (IFN-γ) production was developed. Cells from two subject groups, based on dengue-exposure, were selected for assay development. The unique feature of the IFN-γ ELISPOT assay is the utilization of dendritic cells pulsed with E proteins as antigen presenting cells. IFN-γ production, ranging from 53-513 spot forming units per million peripheral blood mononuclear cells (PBMCs), was observed in dengue-exposed subjects as compared to 0-45 IFN-γ spot forming units in dengue-unexposed subjects. Further, both CD4(+) and CD8(+) T cells, and cells bearing CD45RO memory marker, were the major sources of IFN-γ production. The assay allowed quantification of E-specific IFN-γ-secreting memory T cells in subjects 9 years after exposure to a live-attenuated virus vaccine and live-virus challenge. Results suggested that the dendritic cell-based IFN-γ assay is a useful tool for assessing immunological memory for clinical research.
Collapse
Affiliation(s)
- Peifang Sun
- Henry Jackson Foundation for the Military Service, Rockville, MD, USA.
| | | | | | | | | | | | | |
Collapse
|
415
|
Kusi KA, Faber BW, van der Eijk M, Thomas AW, Kocken CHM, Remarque EJ. Immunization with different PfAMA1 alleles in sequence induces clonal imprint humoral responses that are similar to responses induced by the same alleles as a vaccine cocktail in rabbits. Malar J 2011; 10:40. [PMID: 21320299 PMCID: PMC3050776 DOI: 10.1186/1475-2875-10-40] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Accepted: 02/14/2011] [Indexed: 11/23/2022] Open
Abstract
Background Antibodies to key Plasmodium falciparum surface antigens have been shown to be important effectors that mediate clinical immunity to malaria. The cross-strain fraction of anti-malarial antibodies may however be required to achieve strain-transcending immunity. Such antibody responses against Plasmodium falciparum apical membrane antigen 1 (PfAMA1), a vaccine target molecule that is expressed in both liver and blood stages of the parasite, can be elicited through immunization with a mixture of allelic variants of the parasite molecule. Cross-strain antibodies are most likely elicited against epitopes that are shared by the allelic antigens in the vaccine cocktail. Methods A standard competition ELISA was used to address whether the antibody response can be further focused on shared epitopes by exclusively boosting these common determinants through immunization of rabbits with different PfAMA1 alleles in sequence. The in vitro parasite growth inhibition assay was used to further evaluate the functional effects of the broadened antibody response that is characteristic of multi-allele vaccine strategies. Results A mixed antigen immunization protocol elicited humoral responses that were functionally similar to those elicited by a sequential immunization protocol (p > 0.05). Sequential exposure to the different PfAMA1 allelic variants induced immunological recall of responses to previous alleles and yielded functional cross-strain antibodies that would be capable of optimal growth inhibition of variant parasites at high enough concentrations. Conclusions These findings may have implications for the current understanding of the natural acquisition of clinical immunity to malaria as well as for rational vaccine design.
Collapse
Affiliation(s)
- Kwadwo A Kusi
- Department of Parasitology, Biomedical Primate Research Centre, Postbox 3306, 2280 GH Rijswijk, The Netherlands
| | | | | | | | | | | |
Collapse
|
416
|
Selin LK, Wlodarczyk MF, Kraft AR, Nie S, Kenney LL, Puzone R, Celada F. Heterologous immunity: immunopathology, autoimmunity and protection during viral infections. Autoimmunity 2011; 44:328-47. [PMID: 21250837 DOI: 10.3109/08916934.2011.523277] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Heterologous immunity is a common phenomenon present in all infections. Most of the time it is beneficial, mediating protective immunity, but in some individuals that have the wrong crossreactive response it leads to a cascade of events that result in severe immunopathology. Infections have been associated with autoimmune diseases such as diabetes, multiple sclerosis and lupus erythematosis, but also with unusual autoimmune like pathologies where the immune system appears dysregulated, such as, sarcoidosis, colitis, panniculitis, bronchiolitis obliterans, infectious mononucleosis and even chronic fatigue syndrome. Here we review the evidence that to better understand these autoreactive pathologies it requires an evaluation of how T cells are regulated and evolve during sequential infections with different pathogens under the influence of heterologous immunity.
Collapse
Affiliation(s)
- Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | | | | | | | |
Collapse
|
417
|
Beaumier CM, Jaiswal S, West KY, Friberg H, Mathew A, Rothman AL. Differential in vivo clearance and response to secondary heterologous infections by H2(b)-restricted dengue virus-specific CD8+ T cells. Viral Immunol 2011; 23:477-85. [PMID: 20883162 DOI: 10.1089/vim.2010.0034] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) are hypothesized to play a role in clearance during primary dengue virus (DENV) infections, and contribute to immunopathology during secondary heterologous infections in humans. We previously reported skewed T-cell responses to secondary DENV infection in BALB/c (H-2(d)) mice, reproducing characteristics of human DENV infection. To set the stage for using widely available transgenic and knockout mice, we extended these studies to identify DENV-specific T-cell responses in C57BL/6 (H-2(b)) mice. We identified dominant CD8+ T-cell responses to H-2D(b)-restricted epitopes on the DENV NS4a (aa 249-265) and NS5 (aa 521-537) proteins. High frequencies of IFN-γ- and TNF-α-producing T cells directed at both epitopes were detected following primary infection with all four DENV serotypes, and were augmented by secondary DENV infections. In vivo cytotoxicity assays demonstrated rapid clearance of target cells pulsed with the NS4a peptide; in contrast, NS5 peptide-pulsed target cells were poorly cleared in vivo. These data characterize two H-2(b)-restricted T-cell epitopes displaying divergent in vivo function. These results should facilitate further studies of the in vivo effects of DENV-specific T cells, including the use of genetically modified mouse strains.
Collapse
Affiliation(s)
- Coreen M Beaumier
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | | | |
Collapse
|
418
|
Wang L, Chen RF, Liu JW, Lee IK, Lee CP, Kuo HC, Huang SK, Yang KD. DC-SIGN (CD209) Promoter -336 A/G polymorphism is associated with dengue hemorrhagic fever and correlated to DC-SIGN expression and immune augmentation. PLoS Negl Trop Dis 2011; 5:e934. [PMID: 21245921 PMCID: PMC3014977 DOI: 10.1371/journal.pntd.0000934] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2010] [Accepted: 12/02/2010] [Indexed: 12/29/2022] Open
Abstract
Background The C-type lectin DC-SIGN (CD209) is known to be the major dengue receptor on human dendritic cells, and a single nucleotide polymorphism (SNP) in the promoter region of CD209 (−336 A/G; rs4804803) is susceptible to many infectious diseases. We reason that variations in the DC-SIGN gene might have a broad influence on viral replication and host immune responses. Methods and Findings We studied whether the rs4804803 SNP was associated with a susceptibility to dengue fever (DF) and/or dengue hemorrhagic fever (DHF) through genotyping analysis in a Taiwanese cohort. We generated monocyte-derived dendritic cells (MDDCs) from individuals with AA or AG genotype of rs4804803 to study the viral replication and immune responses for functional validation. A total of 574 DNA samples were genotyped, including 176 DF, 135 DHF, 143 other non-dengue febrile illnesses (OFI) and 120 population controls. A strong association between GG/AG genotypes of rs4804803 and risk of DHF was found when compared among DF, OFI and controls (p = 0.004, 3×10−5 and 0.001, respectively). The AA genotype was associated with protection against dengue infection compared with OFI and controls (p = 0.002 and 0.020, respectively). Moreover, MDDCs from individuals with AG genotype with a higher cell surface DC-SIGN expression had a significantly higher TNFα, IL-12p40, and IP-10 production than those with AA genotype in response to dengue infection. However, the viral replication in MDDCs with AG genotype was significantly lower than those with AA genotype. With both genotypes, MDDCs revealed an increase in viral replication following the addition of anti-IP-10 neutralizing antibody. Conclusions/Significance The rs4804803 SNP in the CD209 promoter contributed to susceptibility to dengue infection and complication of DHF. This SNP with AG genotype affects the cell surface DC-SIGN expression related to immune augmentation and less viral replication. Dengue fever (DF) is an arthropod-borne disease that is prevalent in tropical and subtropical regions of the world. DC-SIGN [dendritic cell-specific intercellular adhesion molecule 3 (ICAM-3)-grabbing non-integrin] is a major receptor for dengue infection. DC-SIGN, also called CD209, expresses on dendritic cells (DCs) that bind to ICAM-3, which is expressed on T cells to facilitate the initial interaction between DCs and T cells. Variations in the CD209 promoter (−336 A/G; rs4804803) genotype are involved in the pathogenesis of human infectious diseases. Here we found that patients with dengue hemorrhagic fever (DHF) had a higher frequency of the AG or GG genotype of rs4804803 than DF or controls. Functional studies determined that monocyte-derived DCs (MDDCs) from individuals with AG genotype had significantly higher cell surface DC-SIGN expression, associated with higher TNFα, IL-12p40, and IP-10 production, but lower viral replication than those with AA genotype. An increase in DEN-2 replication in MDDCs was observed following the addition of anti-IP-10 neutralizing antibody. These findings highlight the fact that the rs4804803 SNP in the CD209 promoter is associated with DHF and correlated to DC-SIGN expression and immune augmentation.
Collapse
Affiliation(s)
- Lin Wang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center (CGMH-KMC), Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Rong-Fu Chen
- Department of Medical Research, CGMH-KMC, Kaohsiung, Taiwan
| | - Jien-Wei Liu
- Division of Infectious Diseases, Department of Internal Medicine, CGMH-KMC, Kaohsiung, Taiwan
| | - Ing-Kit Lee
- Division of Infectious Diseases, Department of Internal Medicine, CGMH-KMC, Kaohsiung, Taiwan
| | - Chiu-Ping Lee
- Department of Medical Research, CGMH-KMC, Kaohsiung, Taiwan
| | - Ho-Chang Kuo
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center (CGMH-KMC), Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shau-Ku Huang
- Johns Hopkins Asthma and Allergy Center, Baltimore, Maryland, United States of America
| | - Kuender D. Yang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center (CGMH-KMC), Kaohsiung, Taiwan
- Department of Medical Research, CGMH-KMC, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- * E-mail:
| |
Collapse
|
419
|
Abstract
OBJECTIVES To provide a comprehensive review of dengue, with an emphasis on clinical syndromes, classification, diagnosis, and management, and to outline relevant aspects of epidemiology, immunopathogenesis, and prevention strategies. Dengue, a leading cause of childhood mortality in Asia and South America, is the most rapidly spreading and important arboviral disease in the world and has a geographic distribution of > 100 countries. DATA SOURCE Boolean searches were carried out by using PubMed from 1975 to March 2009 and the Cochrane Database of Systematic Reviews from 1993 to March 2009 to identify potentially relevant articles by key search terms such as: "dengue"; "dengue fever"; "dengue hemorrhagic fever"; "dengue shock syndrome"; "severe dengue" and "immunopathogenesis," pathogenesis," "classification," "complications," and "management." In addition, authoritative seminal and up-to-date reviews by experts were used. STUDY SELECTION Original research and up-to-date reviews and authoritative reviews consensus statements relevant to diagnosis and therapy were selected. DATA EXTRACTION AND SYNTHESIS We considered the most relevant articles that would be important and of interest to the critical care practitioner as well as authoritative consensus statements from the World Health Organization and the Centers for Disease Control and Prevention. Dengue viral infections are caused by one of four single-stranded ribonucleic acid viruses of the family Flaviviridae and are transmitted by their mosquito vector, Aedes aegypti. The clinical syndromes caused by dengue viral infections occur along a continuum; most cases are asymptomatic and few present with severe forms characterized by shock. Management is predominantly supportive and includes methods to judiciously resolve shock and control bleeding while at the same time preventing fluid overload. CONCLUSIONS Dengue is no longer confined to the tropics and is a global disease. Treatment is supportive. Outcomes can be optimized by early recognition and cautious titrated fluid replacement, especially in resource-limited environments.
Collapse
|
420
|
Midgley CM, Bajwa-Joseph M, Vasanawathana S, Limpitikul W, Wills B, Flanagan A, Waiyaiya E, Tran HB, Cowper AE, Chotiyarnwon P, Grimes JM, Yoksan S, Malasit P, Simmons CP, Mongkolsapaya J, Screaton GR. An in-depth analysis of original antigenic sin in dengue virus infection. J Virol 2011; 85:410-21. [PMID: 20980526 PMCID: PMC3014204 DOI: 10.1128/jvi.01826-10] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 10/14/2010] [Indexed: 01/06/2023] Open
Abstract
The evolution of dengue viruses has resulted in four antigenically similar yet distinct serotypes. Infection with one serotype likely elicits lifelong immunity to that serotype, but generally not against the other three. Secondary or sequential infections are common, as multiple viral serotypes frequently cocirculate. Dengue infection, although frequently mild, can lead to dengue hemorrhagic fever (DHF) which can be life threatening. DHF is more common in secondary dengue infections, implying a role for the adaptive immune response in the disease. There is currently much effort toward the design and implementation of a dengue vaccine but these efforts are made more difficult by the challenge of inducing durable neutralizing immunity to all four viruses. Domain 3 of the dengue virus envelope protein (ED3) has been suggested as one such candidate because it contains neutralizing epitopes and it was originally thought that relatively few cross-reactive antibodies are directed to this domain. In this study, we performed a detailed analysis of the anti-ED3 response in a cohort of patients suffering either primary or secondary dengue infections. The results show dramatic evidence of original antigenic sin in secondary infections both in terms of binding and enhancement activity. This has important implications for dengue vaccine design because heterologous boosting is likely to maintain the immunological footprint of the first vaccination. On the basis of these findings, we propose a simple in vitro enzyme-linked immunosorbent assay (ELISA) to diagnose the original dengue infection in secondary dengue cases.
Collapse
Affiliation(s)
- Claire M. Midgley
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Martha Bajwa-Joseph
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sirijitt Vasanawathana
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Wannee Limpitikul
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Bridget Wills
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Aleksandra Flanagan
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Emily Waiyaiya
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Hai Bac Tran
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Alison E. Cowper
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Pojchong Chotiyarnwon
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Jonathan M. Grimes
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sutee Yoksan
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Prida Malasit
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Cameron P. Simmons
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Juthathip Mongkolsapaya
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Gavin R. Screaton
- Department of Medicine, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom, Paediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khon Kaen, Thailand, Paediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla, Thailand, Oxford University Clinical Research Unit, Wellcome Trust Major Overseas Program, Hospital for Tropical Diseases, Ho Chi Minh City, Viet Nam, Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom, Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand, Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand, Medical Biotechnology Unit, National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| |
Collapse
|
421
|
Friberg H, Burns L, Woda M, Kalayanarooj S, Endy TP, Stephens HA, Green S, Rothman AL, Mathew A. Memory CD8+ T cells from naturally acquired primary dengue virus infection are highly cross-reactive. Immunol Cell Biol 2011; 89:122-9. [PMID: 20421879 PMCID: PMC2929403 DOI: 10.1038/icb.2010.61] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cross-reactive memory T cells induced by primary infection with one of the four serotypes of dengue virus (DENV) are hypothesized to have an immunopathological function in secondary heterologous DENV infection. To define the T-cell response to heterologous serotypes, we isolated HLA-A(*)1101-restricted epitope-specific CD8(+) T-cell lines from primary DENV-immune donors. Cell lines exhibited marked cross-reactivity toward peptide variants representing the four DENV serotypes in tetramer binding and functional assays. Many clones responded similarly to homologous and heterologous serotypes with striking cross-reactivity between the DENV-1 and DENV-3 epitope variants. In vitro-stimulated T-cell lines consistently revealed a hierarchical induction of MIP-1β>degranulation>tumor necrosis factor α (TNFα)>interferon-γ (IFNγ), which depended on the concentration of agonistic peptide. Phosphoflow assays showed peptide dose-dependent phosphorylation of ERK1/2, which correlated with cytolysis, degranulation, and induction of TNFα and IFNγ, but not MIP-1β production. This is the first study to show significant DENV serotype-cross-reactivity of CD8(+) T cells after naturally acquired primary infection. We also show qualitatively different T-cell receptor signaling after stimulation with homologous and heterologous peptides. Our data support a model whereby the order of sequential DENV infections influences the immune response to secondary heterologous DENV infection, contributing to varying disease outcomes.
Collapse
Affiliation(s)
- Heather Friberg
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Lynne Burns
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Marcia Woda
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | | | - Timothy P. Endy
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Henry A.F. Stephens
- Centre for Nephrology and the Anthony Nolan Trust, Royal Free Campus, University College London, UK
| | - Sharone Green
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Alan L. Rothman
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Anuja Mathew
- Center for Infectious Disease and Vaccine Research, University of Massachusetts Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
422
|
Modhiran N, Kalayanarooj S, Ubol S. Subversion of innate defenses by the interplay between DENV and pre-existing enhancing antibodies: TLRs signaling collapse. PLoS Negl Trop Dis 2010; 4:e924. [PMID: 21200427 PMCID: PMC3006139 DOI: 10.1371/journal.pntd.0000924] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Accepted: 11/23/2010] [Indexed: 12/24/2022] Open
Abstract
Background The phenomenon of antibody dependent enhancement as a major determinant that exacerbates disease severity in DENV infections is well accepted. While the detailed mechanism of antibody enhanced disease severity is unclear, evidence suggests that it is associated with both increased DENV infectivity and suppression of the type I IFN and pro-inflammatory cytokine responses. Therefore, it is imperative for us to understand the intracellular mechanisms altered during ADE infection to decipher the mechanism of severe pathogenesis. Methodology/Principal Findings In this present work, qRT-PCR, immunoblotting and gene array analysis were conducted to determine whether DENV-antibody complex infection exerts a suppressive effect on the expression and/or function of the pathogen recognition patterns, focusing on the TLR-signaling pathway. We show here that FcγRI and FcγRIIa synergistically facilitated entry of DENV-antibody complexes into monocytic THP-1 cells. Ligation between DENV-antibody complexes and FcR not only down regulated TLRs gene expression but also up regulated SARM, TANK, and negative regulators of the NF-κB pathway, resulting in suppression of innate responses but increased viral production. These results were confirmed by blocking with anti-FcγRI or anti-FcγRIIa antibodies which reduced viral production, up-regulated IFN-β synthesis, and increased gene expression in the TLR-dependent signaling pathway. The negative impact of DENV-ADE infection on the TLR-dependent pathway was strongly supported by gene array screening which revealed that both MyD88-dependent and –independent signaling molecules were down regulated during DENV-ADE infection. Importantly, the same phenomenon was seen in PBMC of secondary DHF/DSS patients but not in PBMC of DF patients. Conclusions/Significance Our present work demonstrates the mechanism by which DENV uses pre-existing immune mediators to defeat the principal activating pathway of innate defense resulting in suppression of an array of innate immune responses. Interestingly, this phenomenon specifically occurred during the severe form of DENV infection but not in the mild form of disease. Dengue is the most common vector-borne viral disease in humans, with 50–100 million infections per year. The severity of dengue ranges from an acute febrile illness, DF, to a life-threatening vascular leakage syndrome with or without shock, DHF/DSS. Determinants of these syndromes are mainly host factors including non protective but cross reactive antibodies which are known as preexisting enhancing antibodies. These antibodies enhance disease severity through increasing the virus infected cell mass and facilitating intracellular virus replication. Here we demonstrate that DENV exploits preexisting subneutralizing antibodies to defeat the pathogen recognition system and to down regulate the TLR signaling pathway resulting in suppression of an array of innate immune responses. Furthermore, we also show that this phenomenon specifically occurs in the severe form of dengue but not in the mild form of disease.
Collapse
Affiliation(s)
- Naphak Modhiran
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Siripen Kalayanarooj
- WHO Collaborating Centre Case Management of Dengue/DHF/DSS, Queen Sirikit National Institute of Child Health, Bangkok, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
423
|
Yauch LE, Prestwood TR, May MM, Morar MM, Zellweger RM, Peters B, Sette A, Shresta S. CD4+ T cells are not required for the induction of dengue virus-specific CD8+ T cell or antibody responses but contribute to protection after vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:5405-16. [PMID: 20870934 PMCID: PMC2962919 DOI: 10.4049/jimmunol.1001709] [Citation(s) in RCA: 157] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The contribution of T cells to the host response to dengue virus (DENV) infection is not well understood. We previously demonstrated a protective role for CD8(+) T cells during primary DENV infection using a mouse-passaged DENV strain and IFN-α/βR(-/-) C57BL/6 mice, which are susceptible to DENV infection. In this study, we examine the role of CD4(+) T cells during primary DENV infection. Four I-A(b)-restricted epitopes derived from three of the nonstructural DENV proteins were identified. CD4(+) T cells expanded and were activated after DENV infection, with peak activation occurring on day 7. The DENV-specific CD4(+) T cells expressed intracellular IFN-γ, TNF, IL-2, and CD40L, and killed peptide-pulsed target cells in vivo. Surprisingly, depletion of CD4(+) T cells before DENV infection had no effect on viral loads. Consistent with this observation, CD4(+) T cell depletion did not affect the DENV-specific IgG or IgM Ab titers or their neutralizing activity, or the DENV-specific CD8(+) T cell response. However, immunization with the CD4(+) T cell epitopes before infection resulted in significantly lower viral loads. Thus, we conclude that whereas CD4(+) T cells are not required for controlling primary DENV infection, their induction by immunization can contribute to viral clearance. These findings suggest inducing anti-DENV CD4(+) T cell responses by vaccination may be beneficial.
Collapse
Affiliation(s)
- Lauren E. Yauch
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Tyler R. Prestwood
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Monica M. May
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Malika M. Morar
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Raphaël M. Zellweger
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| | - Sujan Shresta
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037
| |
Collapse
|
424
|
Duangchinda T, Dejnirattisai W, Vasanawathana S, Limpitikul W, Tangthawornchaikul N, Malasit P, Mongkolsapaya J, Screaton G. Immunodominant T-cell responses to dengue virus NS3 are associated with DHF. Proc Natl Acad Sci U S A 2010; 107:16922-7. [PMID: 20837518 PMCID: PMC2947904 DOI: 10.1073/pnas.1010867107] [Citation(s) in RCA: 184] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue infections are increasing at an alarming rate in many tropical and subtropical countries, where epidemics can put health care systems under extreme pressure. The more severe infections lead to dengue hemorrhagic fever (DHF), which can be life threatening. A variety of viral and host factors have been associated with the severity of dengue infections. Because secondary dengue infection is more commonly associated with DHF than primary infections, the acquired immune response to dengue, both B cells and T cells have been implicated. In this study, we set out to study T-cell responses across the entire dengue virus proteome and to see whether these were related to disease severity in a cohort of dengue-infected children from Thailand. Robust responses were observed in most infected individuals against most viral proteins. Responses to NS3 were the most frequent, and there was a very strong association between the magnitude of the response and disease severity. Furthermore, in DHF, cytokine-high CD107a-negative cells predominated.
Collapse
Affiliation(s)
- Thaneeya Duangchinda
- Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Wanwisa Dejnirattisai
- Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| | - Sirijit Vasanawathana
- Pediatric Department, Khon Kaen Hospital, Ministry of Public Health, Khonkaen 40000, Thailand
| | - Wannee Limpitikul
- Pediatric Department, Songkhla Hospital, Ministry of Public Health, Songkhla 90100, Thailand; and
| | - Nattaya Tangthawornchaikul
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
| | - Prida Malasit
- Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani 12120, Thailand
- Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Juthathip Mongkolsapaya
- Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
- Medical Molecular Biology Unit, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Gavin Screaton
- Division of Immunology and Inflammation, Department of Medicine, Faculty of Medicine, Hammersmith Hospital, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
425
|
van Panhuis WG, Gibbons RV, Endy TP, Rothman AL, Srikiatkhachorn A, Nisalak A, Burke DS, Cummings DAT. Inferring the serotype associated with dengue virus infections on the basis of pre- and postinfection neutralizing antibody titers. J Infect Dis 2010; 202:1002-10. [PMID: 20738205 DOI: 10.1086/656141] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND Currently, the only tests capable of determining the serotype associated with dengue virus (DENV) infection require sampling during the period of acute viremia. No test can accurately detect the serotype associated with past DENV infections. The standard assay for determination of serotype-specific antibody against DENV is the plaque reduction neutralization test (PRNT), although performance of this test continues to be evaluated. METHODS From a cohort study among schoolchildren in Thailand, PRNT values were determined in serum samples collected before and after infection. A multinomial logistic regression model was used to infer the serotype associated with intercurrent DENV infections. Models were validated based on polymerase chain reaction identification of DENV serotypes. RESULTS The serotype associated with DENV infection inferred by the model corresponded with polymerase chain reaction in 67.6% of cases, and the kappa statistic was 0.479. A model for 35 cases with primary seroconversion correctly identified the DENV serotypes causing infection in 77.1% of cases, compared with 66.9%, using a model for 169 cases with secondary seroconversion. The best model using only postinfection PRNT values correctly inferred the DENV serotype causing infection in 60.3% of cases. CONCLUSIONS A statistical model based on both pre- and postinfection PRNT values can be used to infer the serotype associated with DENV infections in prospective studies and vaccine trials.
Collapse
Affiliation(s)
- Willem G van Panhuis
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | | | | | |
Collapse
|
426
|
Vaughan K, Greenbaum J, Blythe M, Peters B, Sette A. Meta-analysis of all immune epitope data in the Flavivirus genus: inventory of current immune epitope data status in the context of virus immunity and immunopathology. Viral Immunol 2010; 23:259-84. [PMID: 20565291 DOI: 10.1089/vim.2010.0006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A meta-analysis was performed in order to inventory the immune epitope data related to viruses in the genus Flavivirus. Nearly 2000 epitopes were captured from over 130 individual Flavivirus-related references identified from PubMed and reported as of September 2009. This report includes all epitope structures and associated immune reactivity from the past and current literature, including: the epitope distribution among pathogens and related strains, the epitope distribution among different pathogen antigens, the number of epitopes defined in human and animal models of disease, the relationship between epitopes identified in different disease states following natural (or experimental) infection, and data from studies focused on candidate vaccines. We found that the majority of epitopes were defined for dengue virus (DENV) and West Nile virus (WNV). The prominence of DENV and WNV data in the epitope literature is likely a reflection of their overall worldwide impact on human disease, and the lack of vaccines. Conversely, the relatively smaller number of epitopes defined for the other viruses within the genus (yellow fever and Japanese encephalitis virus) most likely reflects the presence of established prophylaxis and/or their more modest impact on morbidity and mortality globally. Through this work we hope to provide useful data to those working in the area of Flavivirus research.
Collapse
Affiliation(s)
- Kerrie Vaughan
- La Jolla Institute of Allergy and Immunology, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
427
|
Zaiss DMW, Boog CJP, van Eden W, Sijts AJAM. Considerations in the design of vaccines that induce CD8 T cell mediated immunity. Vaccine 2010; 28:7716-22. [PMID: 20851090 DOI: 10.1016/j.vaccine.2010.08.101] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 08/27/2010] [Accepted: 08/31/2010] [Indexed: 12/22/2022]
Abstract
The protective capacity of many currently used vaccines is based on induction of neutralizing antibodies. Many pathogens, however, have adapted themselves in different ways to escape antibody-based immune protection. In particular, for those infections against which conventional neutralizing antibody-based vaccinations appear challenging, CD8 T-cells are considered to be promising candidates for vaccine targeting. The design of vaccines that induce robust and long-lasting protective CD8 T-cell responses however imposes new challenges, as many factors such as kinetics and efficiency of antigen-processing and presentation by antigen presenting cells, T-cell repertoire and cytokine environment during T cell priming contribute to the specificity and functionality of CD8 T-cell responses. In the following, we review the most prominent aspects that underlie CD8 T-cell induction and discuss how this knowledge may help to improve the design of efficient CD8 T-cell inducing vaccines.
Collapse
Affiliation(s)
- D M W Zaiss
- Division of Immunology, Faculty of Veterinary Medicine, University of Utrecht, The Netherlands
| | | | | | | |
Collapse
|
428
|
|
429
|
Genotype-specific neutralization and protection by antibodies against dengue virus type 3. J Virol 2010; 84:10630-43. [PMID: 20702644 DOI: 10.1128/jvi.01190-10] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dengue viruses (DENV) comprise a family of related positive-strand RNA viruses that infect up to 100 million people annually. Currently, there is no approved vaccine or therapy to prevent infection or diminish disease severity. Protection against DENV is associated with the development of neutralizing antibodies that recognize the viral envelope (E) protein. Here, with the goal of identifying monoclonal antibodies (MAbs) that can function as postexposure therapy, we generated a panel of 82 new MAbs against DENV-3, including 24 highly neutralizing MAbs. Using yeast surface display, we localized the epitopes of the most strongly neutralizing MAbs to the lateral ridge of domain III (DIII) of the DENV type 3 (DENV-3) E protein. While several MAbs functioned prophylactically to prevent DENV-3-induced lethality in a stringent intracranial-challenge model of mice, only three MAbs exhibited therapeutic activity against a homologous strain when administered 2 days after infection. Remarkably, no MAb in our panel protected prophylactically against challenge by a strain from a heterologous DENV-3 genotype. Consistent with this, no single MAb neutralized efficiently the nine different DENV-3 strains used in this study, likely because of the sequence variation in DIII within and between genotypes. Our studies suggest that strain diversity may limit the efficacy of MAb therapy or tetravalent vaccines against DENV, as neutralization potency generally correlated with a narrowed genotype specificity.
Collapse
|
430
|
Rodenhuis-Zybert IA, Wilschut J, Smit JM. Dengue virus life cycle: viral and host factors modulating infectivity. Cell Mol Life Sci 2010; 67:2773-86. [PMID: 20372965 PMCID: PMC11115823 DOI: 10.1007/s00018-010-0357-z] [Citation(s) in RCA: 293] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 03/08/2010] [Accepted: 03/16/2010] [Indexed: 11/25/2022]
Abstract
Dengue virus (DENV 1-4) represents a major emerging arthropod-borne pathogen. All four DENV serotypes are prevalent in the (sub) tropical regions of the world and infect 50-100 million individuals annually. Whereas the majority of DENV infections proceed asymptomatically or result in self-limited dengue fever, an increasing number of patients present more severe manifestations, such as dengue hemorrhagic fever and dengue shock syndrome. In this review we will give an overview of the infectious life cycle of DENV and will discuss the viral and host factors that are important in controlling DENV infection.
Collapse
Affiliation(s)
- Izabela A. Rodenhuis-Zybert
- Molecular Virology Section, Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Jan Wilschut
- Molecular Virology Section, Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Jolanda M. Smit
- Molecular Virology Section, Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
431
|
Sierra B, Perez AB, Vogt K, Garcia G, Schmolke K, Aguirre E, Alvarez M, Volk HD, Guzman MG. MCP-1 and MIP-1α expression in a model resembling early immune response to dengue. Cytokine 2010; 52:175-83. [PMID: 20650649 DOI: 10.1016/j.cyto.2010.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2009] [Revised: 06/07/2010] [Accepted: 06/19/2010] [Indexed: 10/19/2022]
Abstract
Dengue virus has become endemic in most tropical urban areas throughout the world, and DHF has appeared concomitantly with this expansion. The intensity of dengue virus replication during the early stages of infection could determine clinical outcomes; therefore, it is important to understand the impact of dengue virus infection on the earliest immune defense against microbial infection, which also strongly regulates the adaptive immune responses. This study was aimed at evaluating the expression of the CC-chemokines MIP-1α/CCL3 and MCP-1/CCL2 in peripheral blood leukocytes using an ex vivo model resembling dengue infection in vivo, in subjects with a well characterized dengue immune background, due to the exceptional Cuban epidemiological situation in dengue. The expression of IFNγ, TNFα and IL10 was also evaluated, giving insight about the role of MCP-1 and MIP-1α in the interplay between innate and adaptive immunity. From individuals with different dengue immune background after dengue virus challenge, increased and different expression of the chemokines and cytokines studied was verified in peripheral blood mononuclear cells, thus demonstrating that the previous immunity to a dengue virus serotype has a strong influence on the early immune response after dengue re-infection.
Collapse
Affiliation(s)
- Beatriz Sierra
- Virology Department, PAHO/WHO Collaborating Center for the Study of Dengue and its Vector, Institute of Tropical Medicine Pedro Kouri, Autopista Novia del Mediodia Km. 6 ½, La Lisa, Havana City, Cuba.
| | | | | | | | | | | | | | | | | |
Collapse
|
432
|
Dung NTP, Duyen HTL, Thuy NTV, Ngoc TV, Chau NVV, Hien TT, Rowland-Jones SL, Dong T, Farrar J, Wills B, Simmons CP. Timing of CD8+ T cell responses in relation to commencement of capillary leakage in children with dengue. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:7281-7. [PMID: 20483770 PMCID: PMC4340505 DOI: 10.4049/jimmunol.0903262] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune activation is a feature of dengue hemorrhagic fever (DHF) and CD8+ T cell responses in particular have been suggested as having a role in the vasculopathy that characterizes this disease. By phenotyping CD8+ T cells (CD38+/HLA-DR+, CD38+/Ki-67+, or HLA-DR+/Ki-67+) in serial blood samples from children with dengue, we found no evidence of increased CD8+ T cell activation prior to the commencement of resolution of viremia or hemoconcentration. Investigations with MHC class I tetramers to detect NS3(133-142)-specific CD8+ T cells in two independent cohorts of children suggested the commencement of hemoconcentration and thrombocytopenia in DHF patients generally begins before the appearance of measurable frequencies of NS3(133-142)-specific CD8+ T cells. The temporal mismatch between the appearance of measurable surface activated or NS3(133-142)-specific CD8+ T cells suggests that these cells are sequestered at sites of infection, have phenotypes not detected by our approach, or that other mechanisms independent of CD8+ T cells are responsible for early triggering of capillary leakage in children with DHF.
Collapse
Affiliation(s)
- Nguyen Thi Phuong Dung
- Oxford University Clinical Research Unit and Hospital for Tropical Diseases, Ho Chi Minh City, Vietnam
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
433
|
Zehn D, Turner MJ, Lefrançois L, Bevan MJ. Lack of original antigenic sin in recall CD8(+) T cell responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:6320-6. [PMID: 20439913 PMCID: PMC2982183 DOI: 10.4049/jimmunol.1000149] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In the real world, mice and men are not immunologically naive, having been exposed to numerous antigenic challenges. Prior infections sometimes negatively impact the response to a subsequent infection. This can occur in serial infections with pathogens sharing cross-reactive Ags. At the T cell level it has been proposed that preformed memory T cells, which cross-react with low avidity to epitopes presented in subsequent infections, dampen the response of high-avidity T cells. We investigated this with a series of related MHC class-I restricted Ags expressed by bacterial and viral pathogens. In all cases, we find that high-avidity CD8(+) T cell precursors, either naive or memory, massively expand in secondary cross-reactive infections to dominate the response over low-avidity memory T cells. This holds true even when >10% of the CD8(+) T cell compartment consists of memory T cells that cross-react weakly with the rechallenge ligand. Occasionally, memory cells generated by low-avidity stimulation in a primary infection recognize a cross-reactive epitope with high avidity and contribute positively to the response to a second infection. Taken together, our data show that the phenomenon of original antigenic sin does not occur in all heterologous infections.
Collapse
Affiliation(s)
- Dietmar Zehn
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
- Service of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Swiss Vaccine Research Institute, Lausanne, Switzerland
| | - Michael J. Turner
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
- Immunotherapy Research, Genzyme Corporation, Framingham, MA 01701
| | - Leo Lefrançois
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Michael J. Bevan
- Department of Immunology, Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
434
|
Zhong W, Liu F, Dong L, Lu X, Hancock K, Reinherz EL, Katz JM, Sambhara S. Significant impact of sequence variations in the nucleoprotein on CD8 T cell-mediated cross-protection against influenza A virus infections. PLoS One 2010; 5:e10583. [PMID: 20485501 PMCID: PMC2868023 DOI: 10.1371/journal.pone.0010583] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 04/19/2010] [Indexed: 11/18/2022] Open
Abstract
Background Memory CD8 T cells to influenza A viruses are widely detectable in healthy human subjects and broadly cross-reactive for serologically distinct influenza A virus subtypes. However, it is not clear to what extent such pre-existing cellular immunity can provide cross-subtype protection against novel emerging influenza A viruses. Methodology/Principal Findings We show in the mouse model that naturally occurring sequence variations of the conserved nucleoprotein of the virus significantly impact cross-protection against lethal disease in vivo. When priming and challenge viruses shared identical sequences of the immunodominant, protective NP366/Db epitope, strong cross-subtype protection was observed. However, when they did not share complete sequence identity in this epitope, cross-protection was considerably reduced. Contributions of virus-specific antibodies appeared to be minimal under these circumstances. Detailed analysis revealed that the magnitude of the memory CD8 T cell response triggered by the NP366/Db variants was significantly lower than those triggered by the homologous NP366/Db ligand. It appears that strict specificity of a dominant public TCR to the original NP366/Db ligand may limit the expansion of cross-reactive memory CD8 T cells to the NP366/Db variants. Conclusions/Significance Pre-existing CD8 T cell immunity may provide substantial cross-protection against heterosubtypic influenza A viruses, provided that the priming and the subsequent challenge viruses share the identical sequences of the immunodominant, protective CTL epitopes.
Collapse
Affiliation(s)
- Weimin Zhong
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America.
| | | | | | | | | | | | | | | |
Collapse
|
435
|
Abstract
Immune memory responses to previously encountered pathogens can sometimes alter the immune response to and the course of infection of an unrelated pathogen by a process known as heterologous immunity. This response can lead to enhanced or diminished protective immunity and altered immunopathology. Here, we discuss the nature of T-cell cross-reactivity and describe matrices of epitopes from different viruses eliciting cross-reactive CD8(+) T-cell responses. We examine the parameters of heterologous immunity mediated by these cross-reactive T cells during viral infections in mice and humans. We show that heterologous immunity can disrupt T-cell memory pools, alter the complexity of the T-cell repertoire, change patterns of T-cell immunodominance, lead to the selection of viral epitope-escape variants, alter the pathogenesis of viral infections, and, by virtue of the private specificity of T-cell repertoires within individuals, contribute to dramatic variations in viral disease. We propose that heterologous immunity is an important factor in resistance to and variations of human viral infections and that issues of heterologous immunity should be considered in the design of vaccines.
Collapse
Affiliation(s)
- Raymond M Welsh
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | | | |
Collapse
|
436
|
Flatz L, Rieger T, Merkler D, Bergthaler A, Regen T, Schedensack M, Bestmann L, Verschoor A, Kreutzfeldt M, Brück W, Hanisch UK, Günther S, Pinschewer DD. T cell-dependence of Lassa fever pathogenesis. PLoS Pathog 2010; 6:e1000836. [PMID: 20360949 PMCID: PMC2847900 DOI: 10.1371/journal.ppat.1000836] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 02/26/2010] [Indexed: 12/22/2022] Open
Abstract
Lassa virus (LASV), the causative agent of Lassa fever (LF), is endemic in West Africa, accounting for substantial morbidity and mortality. In spite of ongoing research efforts, LF pathogenesis and mechanisms of LASV immune control remain poorly understood. While normal laboratory mice are resistant to LASV, we report that mice expressing humanized instead of murine MHC class I (MHC-I) failed to control LASV infection and develop severe LF. Infection of MHC-I knockout mice confirmed a key role for MHC-I-restricted T cell responses in controlling LASV. Intriguingly we found that T cell depletion in LASV-infected HHD mice prevented disease, irrespective of high-level viremia. Widespread activation of monocyte/macrophage lineage cells, manifest through inducible NO synthase expression, and elevated IL-12p40 serum levels indicated a systemic inflammatory condition. The absence of extensive monocyte/macrophage activation in T cell-depleted mice suggested that T cell responses contribute to deleterious innate inflammatory reactions and LF pathogenesis. Our observations in mice indicate a dual role for T cells, not only protecting from LASV, but also enhancing LF pathogenesis. The possibility of T cell-driven enhancement and immunopathogenesis should be given consideration in future LF vaccine development. Lassa virus (LASV) is the causative agent of Lassa fever (LF), accounting for substantial morbidity and mortality in West Africa. Yet the mechanisms leading to disease remain poorly understood. Here we propose a concept whereby the body's immune defense either defeats LASV rapidly or, if unsuccessful, becomes an essential facilitator of disease. This latter paradoxical postulate stems from observations in genetically engineered (HHD) mice, which we found to be susceptible to LF. HHD mice differ from resistant wild type mice in that they have a humanized repertoire of T cells, a main component of the mammalian immune system. Counterintuitively, we could protect HHD mice against LF by experimentally removing their T cells. We further found that LF correlated with widespread activation of macrophages, which again depended on T cells. Similar to T cells, macrophages are important players in our body's defense system, but their inflammatory products are also candidate mediators of LF. Taken together, these findings suggest that LF may represent an inappropriate host response to infection. Specifically, our study demonstrates a two-faced role of T cell responses against LASV. Such detrimental aspects of immune defense need to be given consideration in future LF vaccine development, to avoid enhancement of disease in vaccinated individuals.
Collapse
Affiliation(s)
- Lukas Flatz
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Institute of Experimental Immunology, Department of Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Toni Rieger
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Doron Merkler
- Department of Neuropathology, Georg-August-University, Göttingen, Germany
| | - Andreas Bergthaler
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Institute of Experimental Immunology, Department of Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Tommy Regen
- Department of Neuropathology, Georg-August-University, Göttingen, Germany
| | | | - Lukas Bestmann
- Institute of Clinical Chemistry, University Hospital of Zurich, Zurich, Switzerland
- Unilabs Dr. Weber, St. Gallen, Switzerland
| | - Admar Verschoor
- Institute of Experimental Immunology, Department of Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Mario Kreutzfeldt
- Department of Neuropathology, Georg-August-University, Göttingen, Germany
| | - Wolfgang Brück
- Department of Neuropathology, Georg-August-University, Göttingen, Germany
| | | | - Stephan Günther
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg, Germany
| | - Daniel D. Pinschewer
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- Institute of Experimental Immunology, Department of Pathology, University Hospital of Zurich, Zurich, Switzerland
- W.H.O. Collaborating Center for Neonatal Vaccinology, University of Geneva, Geneva, Switzerland
- * E-mail:
| |
Collapse
|
437
|
Nie S, Lin SJ, Kim SK, Welsh RM, Selin LK. Pathological features of heterologous immunity are regulated by the private specificities of the immune repertoire. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2107-12. [PMID: 20348239 DOI: 10.2353/ajpath.2010.090656] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Heterologous immunity associated with cross-reactive T-cell responses is proposed to contribute to variations among individuals in the pathogenesis of human viral infections. In genetically identical mice with similar infection histories, marked variations in the magnitude and specificities of T-cell responses under conditions of heterologous immunity occur and have been linked to the private specificity of T-cell repertoires in individual immune mice. Variations in immunopathology in the form of panniculitis are observed in lymphocytic choriomeningitis virus-immune mice after vaccinia virus infection. By adoptively transferring splenocytes from individual lymphocytic choriomeningitis virus-immune donors into paired recipients, we show here that, on vaccinia virus infection, similar levels of panniculitis were generated in recipients from a single donor, but the severity of panniculitis varied among recipients receiving cells from different donors. This indicates that virus-induced immunopathology under conditions of heterologous immunity is a function of the private specificity of the immune repertoire.
Collapse
Affiliation(s)
- Siwei Nie
- Department of Pathology and Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | | | | | |
Collapse
|
438
|
Clements DE, Coller BAG, Lieberman MM, Ogata S, Wang G, Harada KE, Putnak JR, Ivy JM, McDonell M, Bignami GS, Peters ID, Leung J, Weeks-Levy C, Nakano ET, Humphreys T. Development of a recombinant tetravalent dengue virus vaccine: immunogenicity and efficacy studies in mice and monkeys. Vaccine 2010; 28:2705-15. [PMID: 20097152 PMCID: PMC2837772 DOI: 10.1016/j.vaccine.2010.01.022] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 12/08/2009] [Accepted: 01/13/2010] [Indexed: 10/19/2022]
Abstract
Truncated recombinant dengue virus envelope protein subunits (80E) are efficiently expressed using the Drosophila Schneider-2 (S2) cell expression system. Binding of conformationally sensitive antibodies as well as X-ray crystal structural studies indicate that the recombinant 80E subunits are properly folded native-like proteins. Combining the 80E subunits from each of the four dengue serotypes with ISCOMATRIX adjuvant, an adjuvant selected from a set of adjuvants tested for maximal and long lasting immune responses, results in high titer virus neutralizing antibody responses. Immunization of mice with a mixture of all four 80E subunits and ISCOMATRIX adjuvant resulted in potent virus neutralizing antibody responses to each of the four serotypes. The responses to the components of the tetravalent mixture were equivalent to the responses to each of the subunits administered individually. In an effort to evaluate the potential protective efficacy of the Drosophila expressed 80E, the dengue serotype 2 (DEN2-80E) subunit was tested in both the mouse and monkey challenge models. In both models protection against viral challenge was achieved with low doses of antigen in the vaccine formulation. In non-human primates, low doses of the tetravalent formulation induced good virus neutralizing antibody titers to all four serotypes and protection against challenge with the two dengue virus serotypes tested. In contrast to previous reports, where subunit vaccine candidates have generally failed to induce potent, protective responses, native-like soluble 80E proteins expressed in the Drosophila S2 cells and administered with appropriate adjuvants are highly immunogenic and capable of eliciting protective responses in both mice and monkeys. These results support the development of a dengue virus tetravalent vaccine based on the four 80E subunits produced in the Drosophila S2 cell expression system.
Collapse
Affiliation(s)
| | | | | | - Steven Ogata
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | - Gordon Wang
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | - Kent E. Harada
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | - J. Robert Putnak
- Walter Reed Army Institute for Research, Silver Spring, MD 20910
| | - John M. Ivy
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | | | - Gary S. Bignami
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | - Iain D. Peters
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | - Julia Leung
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| | | | | | - Tom Humphreys
- Hawaii Biotech, Inc., 99–193 Aiea Heights Drive, Aiea, HI 96701
| |
Collapse
|
439
|
Abstract
Dengue virus (DV) infection causes either a benign syndrome, dengue fever, or a severe syndrome, dengue haemorrhagic fever/dengue shock syndrome (DHF/DSS), that is characterized by systemic capillary leakage, thrombocytopaenia and hypovolaemic shock. DHF/DSS occur mainly due to secondary infection by a heterotype DV infection in children and adults but in infants even primary infection by DV causes DHF/DSS. Clinical manifestations of DHF/DSS are more significantly associated with death in infants compared with older children. Vertical transmission of DV and anti-DV IgG has been well reported and is responsible for the pathogenesis of DV disease and its manifestations in infants. The complex pathogenesis of DHF/DSS during primary dengue in infants, with multiple age-related confounding factors, offers unique challenges to investigators. Dengue in infants is not often studied in detail due to practical limitations, but looking at the magnitude of DHF/DSS in infants and the unique opportunities this model provides, there is a need to focus on this problem. This paper reviews existing knowledge on this aspect of DV infection and the challenges it provides.
Collapse
Affiliation(s)
- Amita Jain
- Department of Microbiology, CSM Medical University, Lucknow, India
| | | |
Collapse
|
440
|
Calzavara-Silva CE, Gomes ALV, Maia RCC, Acioli-Santos B, Gil LHVG, Marques ETA. Early molecular markers predictive of dengue hemorrhagic fever. AN ACAD BRAS CIENC 2010; 81:671-7. [PMID: 19893893 DOI: 10.1590/s0001-37652009000400006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Accepted: 07/09/2009] [Indexed: 03/09/2023] Open
Abstract
The management of acute dengue patients during outbreaks is a challenging problem. Most of the dengue fever cases are benign, but some cases develop into a severe and possibly lethal vasculopathy, known as dengue hemorrhagic fever. Early symptoms of dengue and hemorrhagic fever are very similar. An early differential diagnosis is needed to predict which of these two clinical presentations is crucial to proper patient care and public health management. This study evaluates the predictive potential of specific mRNA expression markers of dengue hemorrhagic fever using quantitative real-time PCR assays. Six candidate 'dengue hemorrhagic fever specific signature genes' were evaluated and all showed good correlation among their transcription levels at early days of infection and the later development of severe vasculopathy. The markers selected were able to indicate, at early stages of infection, the evolution of a dengue-infected patient to the severe form of the illness. Despite the fact that these results grant further validation studies, the panel of candidate prognostic markers obtained demonstrated the potential to be useful for clinical use in the form of a fast assay based in blood samples.
Collapse
Affiliation(s)
- Carlos E Calzavara-Silva
- Laboratório de Virologia e Terapia Experimental, Centro de Pesquisa Aggeu Magalhães, CPqAM/FIOCRUZ, Recife, PE, Brasil.
| | | | | | | | | | | |
Collapse
|
441
|
Abstract
Dengue viruses (DENV), a group of four serologically distinct but related flaviviruses, are the cause of one of the most important emerging viral diseases. DENV infections result in a wide spectrum of clinical disease including dengue haemorrhagic fever (DHF), a viral haemorrhagic disease characterised by bleeding and plasma leakage. The characteristic feature of DHF is the transient period of plasma leakage and a haemorrhagic tendency. DHF occurs mostly during a secondary DENV infection. Serotype cross-reactive antibodies and mediators from serotype cross-reactive Dengue-specific T cells have been implicated in the pathogenesis. A complex interaction between virus, host immune response and endothelial cells likely impacts the barrier integrity and functions of endothelial cells leading to plasma leakage. Recently the role of angiogenic factors and the role of dengue virus on endothelial cell transcription and functions have been studied. Insights into the mechanisms that confer protection or cause disease are critical in the development of prophylactic and therapeutic modalities for this important disease.
Collapse
Affiliation(s)
- Anon Srikiatkhachorn
- University of Massachusetts Medical School, Center for Infectious Diseases and Vaccine Research, Worcester, MA 01655-0002, USA.
| |
Collapse
|
442
|
Donaldson EF, Lindesmith LC, Lobue AD, Baric RS. Viral shape-shifting: norovirus evasion of the human immune system. Nat Rev Microbiol 2010; 8:231-41. [PMID: 20125087 PMCID: PMC7097584 DOI: 10.1038/nrmicro2296] [Citation(s) in RCA: 212] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Noroviruses are a major cause of gastroenteritis, and there are currently no vaccines or antiviral treatments available to treat or prevent the >260 million gastroenteritis cases that are reported globally each year. Noroviruses have proven difficult to work with in the laboratory owing to the lack of cell culture systems and animal models, and therefore little is known about the pathogenesis caused by this virus, which has hampered the development of efficacious therapeutics. The norovirus family contains two genogroups (GI and GII) that are most commonly associated with enteric disease in humans, and these genogroups contain more than 25 different genotypes that account for most human norovirus cases. However, outbreaks caused by the GII.4 genotype occur much more frequently than those caused by other genotypes in the GII genogroup, and GI outbreaks occur even less frequently. Although the majority of norovirus outbreaks are caused by the GII.4 genotype, the molecular and biological factors that regulate this disease burden are only partially understood. The GII.4 genotype seems to operate in a similar fashion as influenza virus, whereby evolution of novel immune escape variants allows the virus to escape the predominant memory immune response. By contrast, the prototypic GI.1 noroviruses have remained relatively static over the same time period, evolving variants with identical histo-blood group antigen binding capabilities and similar antigenic properties. The molecular mechanisms governing differential evolution patterns remain a key mystery in the norovirus field. Immunity against noroviruses has been difficult to assess owing to the complex effects of host pre-exposure histories and differential host susceptibility, which is correlated with blood group and secretor status. However, recent work has suggested that the GI and GII genogroups may use different mechanisms to escape immunological memory and that this is perhaps directly related to the plasticity of and complex evolutionary-related sequence information encoded in the P2 subdomain of the capsid protein. The GII genogroup contains more amino acid sequence in the P2 subdomain, which may allow increased capsid plasticity and a tolerance for more amino acid variation or insertions and deletions. This would provide a larger repertoire of sequence targets for natural selection and adaptation to complex environmental selection processes, like herd immunity. By contrast, the GI genogroup contains less sequence information with more conserved, surface-exposed residues that are probably recognized by homologous antibodies as well as antibodies generated against heterologous GI strains. Thus, complex patterns of GI pre-exposure history, antibody cross reactivity and original antigenic sin may facilitate secondary infections of GI strains, whereas antigenic drift and receptor switching allow GII noroviruses, especially GII.4 viruses, to persist in human populations.
Noroviruses are the most common cause of food-borne gastroenteritis worldwide; however, the development of effective vaccines and antiviral therapies has proved to be challenging. In this Review, Baric and colleagues discuss the molecular and structural mechanisms underlying the persistence of noroviruses in human populations. Noroviruses are the most common cause of food-borne gastroenteritis worldwide, and explosive outbreaks frequently occur in community settings, where the virus can immobilize large numbers of infected individuals for 24–48 hours, making the development of effective vaccines and antiviral therapies a priority. However, several challenges have hampered therapeutic design, including: the limitations of cell culture and small-animal model systems; the complex effects of host pre-exposure histories; differential host susceptibility, which is correlated with blood group and secretor status; and the evolution of novel immune escape variants. In this Review, we discuss the molecular and structural mechanisms that facilitate the persistence of noroviruses in human populations.
Collapse
Affiliation(s)
- Eric F Donaldson
- University of North Carolina, Department of Epidemiology, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
443
|
Abstract
Much remains to be learned about the pathogenesis of the different manifestations of dengue virus (DENV) infections in humans. They may range from subclinical infection to dengue fever, dengue hemorrhagic fever (DHF), and eventually dengue shock syndrome (DSS). As both cell tropism and tissue tropism of DENV are considered major determinants in the pathogenesis of dengue, there is a critical need for adequate tropism assays, animal models, and human autopsy data. More than 50 years of research on dengue has resulted in a host of literature, which strongly suggests that the pathogenesis of DHF and DSS involves viral virulence factors and detrimental host responses, collectively resulting in abnormal hemostasis and increased vascular permeability. Differential targeting of specific vascular beds is likely to trigger the localized vascular hyperpermeability underlying DSS. A personalized approach to the study of pathogenesis will elucidate the basis of individual risk for development of DHF and DSS as well as identify the genetic and environmental bases for differences in risk for development of severe disease.
Collapse
|
444
|
Sun GQ, Liu QX, Jin Z, Chakraborty A, Li BL. Influence of infection rate and migration on extinction of disease in spatial epidemics. J Theor Biol 2010; 264:95-103. [PMID: 20085769 DOI: 10.1016/j.jtbi.2010.01.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 12/23/2009] [Accepted: 01/07/2010] [Indexed: 11/29/2022]
Abstract
Extinction of disease can be explained by the patterns of epidemic spreading, yet the underlying causes of extinction are far from being well understood. To reveal a mechanism of disease extinction, a cellular automata model with both birth, death rate and migration is presented. We find that, in single patch, when the infection rate is small or large enough, the disease will disappear for a long time. When the invasion form is in the coexistence of stable spiral and turbulent wave state, the disease will persist. Also, we find that the migration has dual effects on the epidemic spreading. On one hand, in the extinction region of single patch, if the migration rate is large enough, there is a phase transition from the disease free to endemic state in two patches. On the other hand, migration will induce extinction in the regime, which can ensure the persistence of the disease in single patch, due to emergence of anti-phase synchrony. The results obtained well reveal the effect of infection rate and migration on the extinction of the disease, which enriches the finding in the filed of epidemiology and may provide some new ideas to control the disease in the real world.
Collapse
Affiliation(s)
- Gui-Quan Sun
- Department of Mathematics, North University of China, Taiyuan, Shan'xi 030051, People's Republic of China.
| | | | | | | | | |
Collapse
|
445
|
Brown MA, Troyer JL, Pecon-Slattery J, Roelke ME, O'Brien SJ. Genetics and pathogenesis of feline infectious peritonitis virus. Emerg Infect Dis 2010; 15:1445-52. [PMID: 19788813 PMCID: PMC2819880 DOI: 10.3201/eid1509.081573] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Coronavirus sequence analyses demonstrate distinctive circulating strains in natural populations. Feline coronavirus (FCoV) is endemic in feral cat populations and cat colonies, frequently preceding outbreaks of fatal feline infectious peritonitis (FIP). FCoV exhibits 2 biotypes: the pathogenic disease and a benign infection with feline enteric coronavirus (FECV). Uncertainty remains regarding whether genetically distinctive avirulent and virulent forms coexist or whether an avirulent form mutates in vivo, causing FIP. To resolve these alternative hypotheses, we isolated viral sequences from FCoV-infected clinically healthy and sick cats (8 FIP cases and 48 FECV-asymptomatic animals); 735 sequences from 4 gene segments were generated and subjected to phylogenetic analyses. Viral sequences from healthy cats were distinct from sick cats on the basis of genetic distances observed in the membrane and nonstructural protein 7b genes. These data demonstrate distinctive circulating virulent and avirulent strains in natural populations. In addition, 5 membrane protein amino acid residues with functional potential differentiated healthy cats from cats with FIP. These findings may have potential as diagnostic markers for virulent FIP-associated FCoV.
Collapse
Affiliation(s)
- Meredith A Brown
- Laboratory of Genomic Diversity, National Cancer Institute, Frederick, Maryland 21702, USA.
| | | | | | | | | |
Collapse
|
446
|
Simmons CP. Dengue. Infect Dis (Lond) 2010. [DOI: 10.1016/b978-0-323-04579-7.00127-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
447
|
Abstract
The immune system recognizes a myriad of invading pathogens and their toxic products. It does so with a finite repertoire of antibodies and T cell receptors. We here describe theories that quantify the dynamics of the immune system. We describe how the immune system recognizes antigens by searching the large space of receptor molecules. We consider in some detail the theories that quantify the immune response to influenza and dengue fever. We review theoretical descriptions of the complementary evolution of pathogens that occurs in response to immune system pressure. Methods including bioinformatics, molecular simulation, random energy models, and quantum field theory contribute to a theoretical understanding of aspects of immunity.
Collapse
Affiliation(s)
- Michael W Deem
- Department of Bioengineering and Physics, Rice University, Houston, TX 77005, USA.
| | | |
Collapse
|
448
|
Rothman AL. Cellular Immunology of Sequential Dengue Virus Infection and its Role in Disease Pathogenesis. Curr Top Microbiol Immunol 2010; 338:83-98. [DOI: 10.1007/978-3-642-02215-9_7] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
449
|
Abstract
Large case control gene association studies have been performed on cohorts of dengue virus (DENV) infected patients identified in mainland Southeast Asia, South Asia and the Caribbean. Candidate genes that have shown statistically significant associations with DENV disease severity encode HLA molecules, cell receptors for IgG (FcGII), vitamin D and ICAM3 (DCSIGN or CD209), pathogen recognition molecules such as mannose binding lectin (MBL), blood related antigens including ABO and human platelet antigens (HPA1 and HPA2). In ethnic Thais with secondary infections a variety of HLA class I alleles (HLA-A 0203, 0207, A11, B 15, B 44, B 46, B 48, B 51, B 52), DCSIGN promoter polymorphisms and the AB blood group, independently associate with either susceptibility or resistance to dengue fever (DF) and the more severe dengue hemorrhagic fever (DHF). There is also evidence that some HLA associations with disease severity correlate with the DENV serotype inducing secondary infections. Taken together, there is now evidence that allelic variants of multiple gene loci involved in both acquired and innate immune responses contribute significantly to DENV disease outcome and severity. Further analysis of the genetic basis of severe DENV disease in different at risk populations may contribute to the development of new preventative and therapeutic interventions.
Collapse
|
450
|
Mosimann ALP, de Borba L, Bordignon J, Mason PW, Santos CNDD. Construction and characterization of a stable subgenomic replicon system of a Brazilian dengue virus type 3 strain (BR DEN3 290-02). J Virol Methods 2010; 163:147-52. [DOI: 10.1016/j.jviromet.2009.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 08/25/2009] [Accepted: 09/07/2009] [Indexed: 11/17/2022]
|