Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Nature ; 505(7485): 691-5, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24284630

ABSTRACT

The type I interferon (IFN) response protects cells from viral infection by inducing hundreds of interferon-stimulated genes (ISGs), some of which encode direct antiviral effectors. Recent screening studies have begun to catalogue ISGs with antiviral activity against several RNA and DNA viruses. However, antiviral ISG specificity across multiple distinct classes of viruses remains largely unexplored. Here we used an ectopic expression assay to screen a library of more than 350 human ISGs for effects on 14 viruses representing 7 families and 11 genera. We show that 47 genes inhibit one or more viruses, and 25 genes enhance virus infectivity. Comparative analysis reveals that the screened ISGs target positive-sense single-stranded RNA viruses more effectively than negative-sense single-stranded RNA viruses. Gene clustering highlights the cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS, also known as MB21D1) as a gene whose expression also broadly inhibits several RNA viruses. In vitro, lentiviral delivery of enzymatically active cGAS triggers a STING-dependent, IRF3-mediated antiviral program that functions independently of canonical IFN/STAT1 signalling. In vivo, genetic ablation of murine cGAS reveals its requirement in the antiviral response to two DNA viruses, and an unappreciated contribution to the innate control of an RNA virus. These studies uncover new paradigms for the preferential specificity of IFN-mediated antiviral pathways spanning several virus families.


Subject(s)
Immunity, Innate/genetics , Immunity, Innate/immunology , Interferons/immunology , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Viruses/immunology , Animals , Cluster Analysis , DNA Viruses/immunology , DNA Viruses/pathogenicity , Flow Cytometry , Gene Library , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferons/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/genetics , RNA Viruses/immunology , RNA Viruses/pathogenicity , STAT1 Transcription Factor/metabolism , Substrate Specificity , Viruses/classification , Viruses/pathogenicity
2.
PLoS Pathog ; 10(4): e1004086, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743949

ABSTRACT

The type I interferon (IFN) signaling response limits infection of many RNA and DNA viruses. To define key cell types that require type I IFN signaling to orchestrate immunity against West Nile virus (WNV), we infected mice with conditional deletions of the type I IFN receptor (IFNAR) gene. Deletion of the Ifnar gene in subsets of myeloid cells resulted in uncontrolled WNV replication, vasoactive cytokine production, sepsis, organ damage, and death that were remarkably similar to infection of Ifnar-/- mice completely lacking type I IFN signaling. In Mavs-/-×Ifnar-/- myeloid cells and mice lacking both Ifnar and the RIG-I-like receptor adaptor gene Mavs, cytokine production was muted despite high levels of WNV infection. Thus, in myeloid cells, viral infection triggers signaling through MAVS to induce proinflammatory cytokines that can result in sepsis and organ damage. Viral pathogenesis was caused in part by massive complement activation, as liver damage was minimized in animals lacking complement components C3 or factor B or treated with neutralizing anti-C5 antibodies. Disease in Ifnar-/- and CD11c Cre+Ifnarf/f mice also was facilitated by the proinflammatory cytokine TNF-α, as blocking antibodies diminished complement activation and prolonged survival without altering viral burden. Collectively, our findings establish the dominant role of type I IFN signaling in myeloid cells in restricting virus infection and controlling pathological inflammation and tissue injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Receptor, Interferon alpha-beta/metabolism , Sepsis/metabolism , Signal Transduction , West Nile Fever/metabolism , West Nile virus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Complement C3/genetics , Complement C3/metabolism , Complement Factor B/genetics , Complement Factor B/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , Mice, Knockout , Receptor, Interferon alpha-beta/genetics , Sepsis/genetics , Sepsis/pathology , Sepsis/virology , West Nile Fever/genetics , West Nile Fever/pathology
3.
PLoS Pathog ; 10(4): e1004072, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24743696

ABSTRACT

We recently described our most potently neutralizing monoclonal antibody, E106, which protected against lethal Dengue virus type 1 (DENV-1) infection in mice. To further understand its functional properties, we determined the crystal structure of E106 Fab in complex with domain III (DIII) of DENV-1 envelope (E) protein to 2.45 Šresolution. Analysis of the complex revealed a small antibody-antigen interface with the epitope on DIII composed of nine residues along the lateral ridge and A-strand regions. Despite strong virus neutralizing activity of E106 IgG at picomolar concentrations, E106 Fab exhibited a ∼20,000-fold decrease in virus neutralization and bound isolated DIII, E, or viral particles with only a micromolar monovalent affinity. In comparison, E106 IgG bound DENV-1 virions with nanomolar avidity. The E106 epitope appears readily accessible on virions, as neutralization was largely temperature-independent. Collectively, our data suggest that E106 neutralizes DENV-1 infection through bivalent engagement of adjacent DIII subunits on a single virion. The isolation of anti-flavivirus antibodies that require bivalent binding to inhibit infection efficiently may be a rare event due to the unique icosahedral arrangement of envelope proteins on the virion surface.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Dengue Virus , Dengue , Immunoglobulin G , Viral Envelope Proteins , Animals , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Antibody Affinity , Dengue/drug therapy , Dengue/immunology , Dengue Virus/chemistry , Dengue Virus/genetics , Dengue Virus/immunology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Immunoglobulin G/chemistry , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Mice , Protein Structure, Quaternary , Protein Structure, Tertiary , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Virion/chemistry , Virion/genetics , Virion/immunology
5.
J Virol ; 88(19): 11007-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25031348

ABSTRACT

UNLABELLED: Upon activation of Toll-like and RIG-I-like receptor signaling pathways, the transcription factor IRF5 translocates to the nucleus and induces antiviral immune programs. The recent discovery of a homozygous mutation in the immunoregulatory gene guanine exchange factor dedicator of cytokinesis 2 (Dock2mu/mu) in several Irf5-/- mouse colonies has complicated interpretation of immune functions previously ascribed to IRF5. To define the antiviral functions of IRF5 in vivo, we infected backcrossed Irf5-/-×Dock2wt/wt mice (here called Irf5-/- mice) and independently generated CMV-Cre Irf5fl/fl mice with West Nile virus (WNV), a pathogenic neurotropic flavivirus. Compared to congenic wild-type animals, Irf5-/- and CMV-Cre Irf5fl/fl mice were more vulnerable to WNV infection, and this phenotype was associated with increased infection in peripheral organs, which resulted in higher virus titers in the central nervous system. The loss of IRF5, however, was associated with only small differences in the type I interferon response systemically and in the draining lymph node during WNV infection. Instead, lower levels of several other proinflammatory cytokines and chemokines, as well as fewer and less activated immune cells, were detected in the draining lymph node 2 days after WNV infection. WNV-specific antibody responses in Irf5-/- mice also were blunted in the context of live or inactivated virus infection and this was associated with fewer antigen-specific memory B cells and long-lived plasma cells. Our results with Irf5-/- mice establish a key role for IRF5 in shaping the early innate immune response in the draining lymph node, which impacts the spread of virus infection, optimal B cell immunity, and disease pathogenesis. IMPORTANCE: Although the roles of IRF3 and IRF7 in orchestrating innate and adaptive immunity after viral infection are established, the function of the related transcription factor IRF5 remains less certain. Prior studies in Irf5-/- mice reported conflicting results as to the contribution of IRF5 in regulating type I interferon and adaptive immune responses. The lack of clarity may stem from a recently discovered homozygous loss-of-function mutation of the immunoregulatory gene Dock2 in several colonies of Irf5-/- mice. Here, using a mouse model with a deficiency in IRF5 and wild-type Dock2 alleles, we investigated how IRF5 modulates West Nile virus (WNV) pathogenesis and host immune responses. Our in vivo studies indicate that IRF5 has a key role in shaping the early proinflammatory cytokine response in the draining lymph node, which impacts immunity and control of WNV infection.


Subject(s)
Antibodies, Viral/blood , Immunity, Innate , Interferon Regulatory Factors/immunology , Lymph Nodes/immunology , West Nile Fever/immunology , West Nile virus/immunology , Adaptive Immunity , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , B-Lymphocytes/virology , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/virology , Crosses, Genetic , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/immunology , Gene Deletion , Gene Expression/immunology , Guanine Nucleotide Exchange Factors , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Mice , Mice, Transgenic , Mutation , Viral Load , West Nile Fever/genetics , West Nile Fever/virology
6.
J Virol ; 87(15): 8363-71, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23740986

ABSTRACT

Previous studies have demonstrated that type I interferon (IFN-I) restricts West Nile virus (WNV) replication and pathogenesis in peripheral and central nervous system (CNS) tissues. However, the in vivo role of specific antiviral genes that are induced by IFN-I against WNV infection remains less well characterized. Here, using Ifit2(-/-) mice, we defined the antiviral function of the interferon-stimulated gene (ISG) Ifit2 in limiting infection and disease in vivo by a virulent North American strain of WNV. Compared to congenic wild-type controls, Ifit2(-/-) mice showed enhanced WNV infection in a tissue-restricted manner, with preferential replication in the CNS of animals lacking Ifit2. Virological analysis of cultured macrophages, dendritic cells, fibroblasts, cerebellar granule cell neurons, and cortical neurons revealed cell type-specific antiviral functions of Ifit2 against WNV. In comparison, small effects of Ifit2 were observed on the induction or magnitude of innate or adaptive immune responses. Our results suggest that Ifit2 restricts WNV infection and pathogenesis in different tissues in a cell type-specific manner.


Subject(s)
Brain/immunology , Brain/virology , Proteins/metabolism , West Nile Fever/immunology , West Nile virus/immunology , Animals , Apoptosis Regulatory Proteins , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteins/genetics , RNA-Binding Proteins , West Nile Fever/pathology , West Nile Fever/virology
7.
J Virol ; 87(21): 11401-15, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23966390

ABSTRACT

Many viruses induce type I interferon responses by activating cytoplasmic RNA sensors, including the RIG-I-like receptors (RLRs). Although two members of the RLR family, RIG-I and MDA5, have been implicated in host control of virus infection, the relative role of each RLR in restricting pathogenesis in vivo remains unclear. Recent studies have demonstrated that MAVS, the adaptor central to RLR signaling, is required to trigger innate immune defenses and program adaptive immune responses, which together restrict West Nile virus (WNV) infection in vivo. In this study, we examined the specific contribution of MDA5 in controlling WNV in animals. MDA5(-/-) mice exhibited enhanced susceptibility, as characterized by reduced survival and elevated viral burden in the central nervous system (CNS) at late times after infection, even though small effects on systemic type I interferon response or viral replication were observed in peripheral tissues. Intracranial inoculation studies and infection experiments with primary neurons ex vivo revealed that an absence of MDA5 did not impact viral infection in neurons directly. Rather, subtle defects were observed in CNS-specific CD8(+) T cells in MDA5(-/-) mice. Adoptive transfer into recipient MDA5(+/+) mice established that a non-cell-autonomous deficiency of MDA5 was associated with functional defects in CD8(+) T cells, which resulted in a failure to clear WNV efficiently from CNS tissues. Our studies suggest that MDA5 in the immune priming environment shapes optimal CD8(+) T cell activation and subsequent clearance of WNV from the CNS.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , DEAD-box RNA Helicases/immunology , Receptors, Pattern Recognition/immunology , West Nile Fever/immunology , West Nile virus/immunology , Animals , CD8-Positive T-Lymphocytes/virology , Central Nervous System/virology , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Interferon-Induced Helicase, IFIH1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Pattern Recognition/metabolism , Survival Analysis , Viral Load , West Nile Fever/virology , West Nile virus/isolation & purification
8.
J Virol ; 87(16): 8826-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23785205

ABSTRACT

Although prior studies have characterized the neutralizing activities of monoclonal antibodies (MAbs) against dengue virus (DENV) serotypes 1, 2, and 3 (DENV-1, DENV-2, and DENV-3), few reports have assessed the activity of MAbs against DENV-4. Here, we evaluated the inhibitory activity of 81 new mouse anti-DENV-4 MAbs. We observed strain- and genotype-dependent differences in neutralization of DENV-4 by MAbs mapping to epitopes on domain II (DII) and DIII of the envelope (E) protein. Several anti-DENV-4 MAbs inefficiently inhibited at least one strain and/or genotype, suggesting that the exposure or sequence of neutralizing epitopes varies within isolates of this serotype. Remarkably, flavivirus cross-reactive MAbs, which bound to the highly conserved fusion loop in DII and inhibited infection of DENV-1, DENV-2, and DENV-3, more weakly neutralized five different DENV-4 strains encompassing the genetic diversity of the serotype after preincubation at 37°C. However, increasing the time of preincubation at 37°C or raising the temperature to 40°C enhanced the potency of DII fusion loop-specific MAbs and some DIII-specific MAbs against DENV-4 strains. Prophylaxis studies in two new DENV-4 mouse models showed that neutralization titers of MAbs after preincubation at 37°C correlated with activity in vivo. Our studies establish the complexity of MAb recognition against DENV-4 and suggest that differences in epitope exposure relative to other DENV serotypes affect antibody neutralization and protective activity.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Animals , Cross Reactions , Dengue/prevention & control , Disease Models, Animal , Epitope Mapping , Mice , Mice, Inbred C57BL , Protein Binding , Temperature , Time Factors
9.
PLoS Pathog ; 8(10): e1002930, 2012.
Article in English | MEDLINE | ID: mdl-23055922

ABSTRACT

We previously developed a panel of neutralizing monoclonal antibodies against Dengue virus (DENV)-1, of which few exhibited inhibitory activity against all DENV-1 genotypes. This finding is consistent with reports observing variable neutralization of different DENV strains and genotypes using serum from individuals that experienced natural infection or immunization. Herein, we describe the crystal structures of DENV1-E111 bound to a novel CC' loop epitope on domain III (DIII) of the E protein from two different DENV-1 genotypes. Docking of our structure onto the available cryo-electron microscopy models of DENV virions revealed that the DENV1-E111 epitope was inaccessible, suggesting that this antibody recognizes an uncharacterized virus conformation. While the affinity of binding between DENV1-E111 and DIII varied by genotype, we observed limited correlation with inhibitory activity. Instead, our results support the conclusion that potent neutralization depends on genotype-dependent exposure of the CC' loop epitope. These findings establish new structural complexity of the DENV virion, which may be relevant for the choice of DENV strain for induction or analysis of neutralizing antibodies in the context of vaccine development.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Dengue Virus/immunology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/immunology , Binding Sites, Antibody , Dengue/immunology , Dengue/prevention & control , Dengue Virus/genetics , Epitope Mapping , Epitopes/genetics , Epitopes/immunology , Humans , Protein Structure, Tertiary
10.
J Virol ; 86(17): 8937-48, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22740407

ABSTRACT

Previous studies of mice have demonstrated that an orchestrated sequence of innate and adaptive immune responses is required to control West Nile virus (WNV) infection in peripheral and central nervous system (CNS) tissues. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL; also known as CD253) has been reported to inhibit infection with dengue virus, a closely related flavivirus, in cell culture. To determine the physiological function of TRAIL in the context of flavivirus infection, we compared the pathogenesis of WNV in wild-type and TRAIL(-/-) mice. Mice lacking TRAIL showed increased vulnerability and death after subcutaneous WNV infection. Although no difference in viral burden was detected in peripheral tissues, greater viral infection was detected in the brain and spinal cord at late times after infection, and this was associated with delayed viral clearance in the few surviving TRAIL(-/-) mice. While priming of adaptive B and T cell responses and trafficking of immune and antigen-specific cells to the brain were undistinguishable from those in normal mice, in TRAIL(-/-) mice, CD8(+) T cells showed qualitative defects in the ability to clear WNV infection. Adoptive transfer of WNV-primed wild-type but not TRAIL(-/-) CD8(+) T cells to recipient CD8(-/-) mice efficiently limited infection in the brain and spinal cord, and analogous results were obtained when wild-type or TRAIL(-/-) CD8(+) T cells were added to WNV-infected primary cortical neuron cultures ex vivo. Collectively, our results suggest that TRAIL produced by CD8(+) T cells contributes to disease resolution by helping to clear WNV infection from neurons in the central nervous system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Neurons/virology , TNF-Related Apoptosis-Inducing Ligand/immunology , West Nile Fever/immunology , West Nile virus/pathogenicity , Adoptive Transfer , Animals , Antibodies, Viral/immunology , Brain/immunology , Brain/virology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/immunology , Spinal Cord/immunology , Spinal Cord/virology , TNF-Related Apoptosis-Inducing Ligand/genetics , Viral Load , West Nile Fever/virology , West Nile virus/immunology , West Nile virus/physiology
11.
PLoS Pathog ; 6(4): e1000823, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20369024

ABSTRACT

Antibody protection against flaviviruses is associated with the development of neutralizing antibodies against the viral envelope (E) protein. Prior studies with West Nile virus (WNV) identified therapeutic mouse and human monoclonal antibodies (MAbs) that recognized epitopes on domain III (DIII) of the E protein. To identify an analogous panel of neutralizing antibodies against DENV type-1 (DENV-1), we immunized mice with a genotype 2 strain of DENV-1 virus and generated 79 new MAbs, 16 of which strongly inhibited infection by the homologous virus and localized to DIII. Surprisingly, only two MAbs, DENV1-E105 and DENV1-E106, retained strong binding and neutralizing activity against all five DENV-1 genotypes. In an immunocompromised mouse model of infection, DENV1-E105 and DENV1-E106 exhibited therapeutic activity even when administered as a single dose four days after inoculation with a heterologous genotype 4 strain of DENV-1. Using epitope mapping and X-ray crystallographic analyses, we localized the neutralizing determinants for the strongly inhibitory MAbs to distinct regions on DIII. Interestingly, sequence variation in DIII alone failed to explain disparities in neutralizing potential of MAbs among different genotypes. Overall, our experiments define a complex structural epitope on DIII of DENV-1 that can be recognized by protective antibodies with therapeutic potential.


Subject(s)
Antibodies, Neutralizing/immunology , Dengue Virus/genetics , Dengue Virus/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes, B-Lymphocyte/immunology , Genotype , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Polymerase Chain Reaction , Protein Structure, Tertiary , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
12.
J Exp Med ; 198(12): 1853-62, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14662909

ABSTRACT

In humans, the elderly and immunocompromised are at greatest risk for disseminated West Nile virus (WNV) infection, yet the immunologic basis for this remains unclear. We demonstrated previously that B cells and IgG contributed to the defense against disseminated WNV infection (Diamond, M.S., B. Shrestha, A. Marri, D. Mahan, and M. Engle. 2003. J. Virol. 77:2578-2586). In this paper, we addressed the function of IgM in controlling WNV infection. C57BL/6J mice (sIgM-/-) that were deficient in the production of secreted IgM but capable of expressing surface IgM and secreting other immunoglobulin isotypes were vulnerable to lethal infection, even after inoculation with low doses of WNV. Within 96 h, markedly higher levels of infectious virus were detected in the serum of sIgM-/- mice compared with wild-type mice. The enhanced viremia correlated with higher WNV burdens in the central nervous system, and was also associated with a blunted anti-WNV IgG response. Passive transfer of polyclonal anti-WNV IgM or IgG protected sIgM-/- mice against mortality, although administration of comparable amounts of a nonneutralizing monoclonal anti-WNV IgM provided no protection. In a prospective analysis, a low titer of anti-WNV IgM antibodies at day 4 uniformly predicted mortality in wild-type mice. Thus, the induction of a specific, neutralizing IgM response early in the course of WNV infection limits viremia and dissemination into the central nervous system, and protects against lethal infection.


Subject(s)
Antibodies, Viral/physiology , Immunoglobulin M/physiology , West Nile Fever/immunology , Animals , Kidney/virology , Mice , Mice, Inbred C57BL , Spleen/virology , Viral Load , Viremia/immunology , West Nile Fever/virology
13.
J Virol ; 82(18): 8956-64, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18632856

ABSTRACT

West Nile virus (WNV) is a neurotropic flavivirus that has emerged globally as a significant cause of viral encephalitis in humans, especially in immunocompromised individuals. Previous studies have shown essential protective roles for antiviral cytokines (e.g., alpha interferon [IFN-alpha] and IFN-gamma) against WNV in mice. However, studies using cell culture offer conflicting answers regarding whether tumor necrosis factor alpha (TNF-alpha) has an anti-WNV function. To test the biological significance of TNF-alpha against WNV in vivo, experiments were performed with TNF receptor-1 (TNF-R1)-deficient and TNF-alpha-depleted C57BL/6 mice. TNF-R1(-/-) mice had enhanced mortality and decreased survival time after WNV infection compared to congenic wild-type mice. Consistent with this, administration of a neutralizing anti-TNF-alpha monoclonal antibody also decreased survival after WNV infection. Relatively small differences in viral burdens in peripheral tissues of TNF-R1(-/-) mice were observed, and this occurrence correlated with a modest antiviral effect of TNF-alpha on primary macrophages but not dendritic cells. In contrast, the viral titers detected in the central nervous systems of TNF-R1(-/-) mice were significantly increased compared to those of wild-type mice, although TNF-alpha did not have a direct antiviral effect in primary neuron cultures. Whereas no defect in priming of adaptive B- and T-cell responses in TNF-R1(-/-) mice was observed, there were significant reductions in accumulations of CD8+ T cells and macrophages in the brain. Our data are most consistent with a model in which interaction of TNF-alpha with TNF-R1 protects against WNV infection by regulating migration of protective inflammatory cells into the brain during acute infection.


Subject(s)
Cell Movement/immunology , Central Nervous System/immunology , Leukocytes, Mononuclear/immunology , Tumor Necrosis Factor-alpha/metabolism , West Nile Fever/immunology , West Nile virus/pathogenicity , Animals , Antibodies, Viral/blood , Brain/immunology , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/virology , Humans , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/genetics , West Nile Fever/mortality , West Nile Fever/virology , West Nile virus/immunology
14.
Nat Med ; 21(12): 1464-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26523970

ABSTRACT

The TAM receptors Tyro3, Axl and Mertk are receptor tyrosine kinases that dampen host innate immune responses following engagement with their ligands Gas6 and Protein S, which recognize phosphatidylserine on apoptotic cells. In a form of apoptotic mimicry, many enveloped viruses display phosphatidylserine on the outer leaflet of their membranes, enabling TAM receptor activation and downregulation of antiviral responses. Accordingly, we hypothesized that a deficiency of TAM receptors would enhance antiviral responses and protect against viral infection. Unexpectedly, mice lacking Mertk and/or Axl, but not Tyro3, exhibited greater vulnerability to infection with neuroinvasive West Nile and La Crosse encephalitis viruses. This phenotype was associated with increased blood-brain barrier permeability, which enhanced virus entry into and infection of the brain. Activation of Mertk synergized with interferon-ß to tighten cell junctions and prevent virus transit across brain microvascular endothelial cells. Because TAM receptors restrict pathogenesis of neuroinvasive viruses, these findings have implications for TAM antagonists that are currently in clinical development.


Subject(s)
Blood-Brain Barrier/enzymology , Blood-Brain Barrier/virology , Encephalitis, California/enzymology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , West Nile Fever/enzymology , Adaptive Immunity , Animals , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/pathology , Chemokines/blood , Encephalitis, California/pathology , Encephalitis, California/virology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Interferon-beta/metabolism , La Crosse virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Permeability , Protective Agents , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/deficiency , Radiation Tolerance , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/deficiency , Signal Transduction , Survival Analysis , Viral Load , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/physiology , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
15.
Viral Immunol ; 16(3): 259-78, 2003.
Article in English | MEDLINE | ID: mdl-14583143

ABSTRACT

WNV continues to spread throughout the Western Hemisphere as virus activity in insects and animals has been reported in the United States, Canada, Mexico, and the Caribbean islands. West Nile virus (WNV) infects the central nervous system and causes severe disease primarily in humans who are immunocompromised or elderly. In this review, we discuss the mechanisms by which the immune system limits dissemination of WNV infection. Recent experimental studies in animals suggest important roles for both the innate and the adaptive immune responses in controlling WNV infection. Interferons, antibody, complement components and CD8+ T cells coordinate protection against severe infection and disease. These findings are analyzed in the context of recent approaches to vaccine development and immunotherapy against WNV.


Subject(s)
West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antibodies, Viral/biosynthesis , Complement Activation , Dendritic Cells/immunology , Humans , Immunity, Cellular , Immunity, Innate , Immunotherapy , In Vitro Techniques , Interferons/biosynthesis , Killer Cells, Natural/immunology , Macrophages/immunology , Models, Immunological , T-Lymphocytes/immunology , Viral Vaccines/isolation & purification , West Nile Fever/therapy , West Nile virus/growth & development , West Nile virus/pathogenicity
16.
Cell Host Microbe ; 14(2): 136-47, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23954153

ABSTRACT

Upon activation by the ligands Gas6 and Protein S, Tyro3/Axl/Mer (TAM) receptor tyrosine kinases promote phagocytic clearance of apoptotic cells and downregulate immune responses initiated by Toll-like receptors and type I interferons (IFNs). Many enveloped viruses display the phospholipid phosphatidylserine on their membranes, through which they bind Gas6 and Protein S and engage TAM receptors. We find that ligand-coated viruses activate TAM receptors on dendritic cells (DCs), dampen type I IFN signaling, and thereby evade host immunity and promote infection. Upon virus challenge, TAM-deficient DCs display type I IFN responses that are elevated in comparison to wild-type cells. As a consequence, TAM-deficient DCs are relatively resistant to infection by flaviviruses and pseudotyped retroviruses, but infection can be restored with neutralizing type I IFN antibodies. Correspondingly, a TAM kinase inhibitor antagonizes the infection of wild-type DCs. Thus, TAM receptors are engaged by viruses in order to attenuate type I IFN signaling and represent potential therapeutic targets.


Subject(s)
Dendritic Cells/immunology , Flavivirus/immunology , Immune Tolerance , Immunity, Innate , Retroviridae/immunology , Animals , Cell Line , Humans , Interferon Type I/biosynthesis , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , c-Mer Tyrosine Kinase , Axl Receptor Tyrosine Kinase
17.
Virology ; 427(2): 127-34, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22406169

ABSTRACT

DENV1-E106 is a monoclonal antibody (MAb) with strong neutralizing activity against all five DENV-1 genotypes and therapeutic activity in mice. Here, we evaluated the potential for DENV-1 to escape neutralization by DENV1-E106. A single mutation in domain III of the envelope protein (T329A) emerged, which conferred resistance to DENV1-E106. However, the T329A variant virus had differing phenotypes in vitro and in vivo with attenuation in cell culture yet increased infectivity in Aedes aegypti mosquitoes. Mice infected with this T329A variant still were protected against lethal infection by DENV1-E106 even though much of the neutralizing activity was lost. This study reveals the complex dynamics of neutralization escape of an inhibitory MAb against DENV, and suggests that evaluation of therapeutic MAbs requires detailed investigation in relevant hosts.


Subject(s)
Aedes/virology , Dengue Virus/immunology , Dengue/virology , Animals , Antibodies, Monoclonal , Cell Line , Cricetinae , Dengue/immunology , Dengue Virus/classification , Dengue Virus/genetics , Dengue Virus/pathogenicity , Insect Vectors , Mice , Mice, Knockout , Mutation , Phenotype , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Virulence
18.
Cell Host Microbe ; 11(4): 397-409, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22520467

ABSTRACT

Host resistance to viral infection requires type I (α/ß) and II (γ) interferon (IFN) production. Another important defense mechanism is the degradative activity of macroautophagy (herein autophagy), mediated by the coordinated action of evolutionarily conserved autophagy proteins (Atg). We show that the Atg5-Atg12/Atg16L1 protein complex, whose prior known function is in autophagosome formation, is required for IFNγ-mediated host defense against murine norovirus (MNV) infection. Importantly, the direct antiviral activity of IFNγ against MNV in macrophages required Atg5-Atg12, Atg7, and Atg16L1, but not induction of autophagy, the degradative activity of lysosomal proteases, fusion of autophagosomes and lysosomes, or the Atg8-processing protein Atg4B. IFNγ, via Atg5-Atg12/Atg16L1, inhibited formation of the membranous cytoplasmic MNV replication complex, where Atg16L1 localized. Thus, the Atg5-Atg12/Atg16L1 complex performs a pivotal, nondegradative role in IFNγ-mediated antiviral defense, establishing that multicellular organisms have evolved to use portions of the autophagy pathway machinery in a cassette-like fashion for host defense.


Subject(s)
Caliciviridae Infections/metabolism , Carrier Proteins/metabolism , Interferon-gamma/immunology , Microtubule-Associated Proteins/metabolism , Norovirus/physiology , Proteins/metabolism , Animals , Autophagy , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Caliciviridae Infections/genetics , Caliciviridae Infections/immunology , Carrier Proteins/genetics , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Protein Binding , Proteins/genetics
19.
Vaccine ; 26(16): 2020-33, 2008 Apr 07.
Article in English | MEDLINE | ID: mdl-18339459

ABSTRACT

West Nile virus (WNV) is a mosquito borne, neurotropic flavivirus that causes a severe central nervous system (CNS) infection in humans and animals. Although commercial vaccines are available for horses, none is currently approved for human use. In this study, we evaluated the efficacy and mechanism of immune protection of two candidate WNV vaccines in mice. A formalin-inactivated WNV vaccine induced higher levels of specific and neutralizing antibodies compared to a DNA plasmid vaccine that produces virus-like particles. Accordingly, partial and almost complete protection against a highly stringent lethal intracranial WNV challenge were observed in mice 60 days after single dose immunization with the DNA plasmid and inactivated virus vaccines, respectively. In mice immunized with a single dose of DNA plasmid or inactivated vaccine, antigen-specific CD8(+) T cells were induced and contributed to protective immunity as acquired or genetic deficiencies of CD8(+) T cells lowered the survival rates. In contrast, in boosted animals, WNV-specific antibody titers were higher, survival rates after challenge were greater, and an absence of CD8(+) T cells did not appreciably affect mortality. Overall, our experiments suggest that in mice, both inactivated WNV and DNA plasmid vaccines are protective after two doses, and the specific contribution of antibody and CD8(+) T cells to vaccine immunity against WNV is modulated by the prime-boost strategy.


Subject(s)
Antibodies, Viral/blood , CD8-Positive T-Lymphocytes/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile virus/immunology , Animals , Antibodies, Viral/immunology , Antibody Specificity , CD8 Antigens/genetics , Immunization, Secondary , Injections, Intramuscular , Injections, Intraperitoneal , Mice , Mice, Congenic , Mice, Inbred C57BL , Neutralization Tests , Plasmids , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , West Nile Fever/blood
20.
J Exp Med ; 205(10): 2319-37, 2008 Sep 29.
Article in English | MEDLINE | ID: mdl-18779347

ABSTRACT

In a lethal West Nile virus (WNV) model, central nervous system infection triggered a threefold increase in CD45(int)/CD11b(+)/CD11c(-) microglia at days 6-7 postinfection (p.i.). Few microglia were proliferating, suggesting that the increased numbers were derived from a migratory precursor cell. Depletion of "circulating" (Gr1(-)(Ly6C(lo))CX3CR1(+)) and "inflammatory" (Gr1(hi)/Ly6C(hi)/CCR2(+)) classical monocytes during infection abrogated the increase in microglia. C57BL/6 chimeras reconstituted with cFMS-enhanced green fluorescent protein (EGFP) bone marrow (BM) showed large numbers of peripherally derived (GFP(+)) microglia expressing GR1(+)(Ly6C(+)) at day 7 p.i., suggesting that the inflammatory monocyte is a microglial precursor. This was confirmed by adoptive transfer of labeled BM (Ly6C(hi)/CD115(+)) or circulating inflammatory monocytes that trafficked to the WNV-infected brain and expressed a microglial phenotype. CCL2 is a chemokine that is highly expressed during WNV infection and important in inflammatory monocyte trafficking. Neutralization of CCL2 not only reduced the number of GFP(+) microglia in the brain during WNV infection but prolonged the life of infected animals. Therefore, CCL2-dependent inflammatory monocyte migration is critical for increases in microglia during WNV infection and may also play a pathogenic role during WNV encephalitis.


Subject(s)
Antigens, Ly/immunology , Brain/immunology , Brain/virology , Microglia/immunology , Monocytes/immunology , Stem Cells/immunology , West Nile Fever/immunology , Adoptive Transfer , Animals , Blood-Brain Barrier/physiology , Brain/cytology , Brain/pathology , Chemokine CCL2/immunology , Clodronic Acid/metabolism , Female , Leukocyte Common Antigens/immunology , Liposomes/chemistry , Liposomes/metabolism , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/physiology , Monocytes/cytology , Stem Cells/cytology , Survival Rate , West Nile Fever/pathology , West Nile virus
SELECTION OF CITATIONS
SEARCH DETAIL