Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Commun Biol ; 7(1): 494, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658802

ABSTRACT

Inflammatory monocytes (iMO) are recruited from the bone marrow to the brain during viral encephalitis. C-C motif chemokine receptor (CCR) 2 deficiency substantially reduces iMO recruitment for most, but not all encephalitic viruses. Here we show CCR7 acts synergistically with CCR2 to control this process. Following Herpes simplex virus type-1 (HSV-1), or La Crosse virus (LACV) infection, we find iMO proportions are reduced by approximately half in either Ccr2 or Ccr7 knockout mice compared to control mice. However, Ccr2/Ccr7 double knockouts eliminate iMO recruitment following infection with either virus, indicating these receptors together control iMO recruitment. We also find that LACV induces a more robust iMO recruitment than HSV-1. However, unlike iMOs in HSV-1 infection, LACV-recruited iMOs do not influence neurological disease development. LACV-induced iMOs have higher expression of proinflammatory and proapoptotic but reduced mitotic, phagocytic and phagolysosomal transcripts compared to HSV-1-induced iMOs. Thus, virus-specific activation of iMOs affects their recruitment, activation, and function.


Subject(s)
Brain , Herpesvirus 1, Human , La Crosse virus , Mice, Knockout , Monocytes , Receptors, CCR2 , Receptors, CCR7 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Mice , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Brain/virology , Brain/metabolism , Brain/immunology , Herpesvirus 1, Human/physiology , La Crosse virus/genetics , La Crosse virus/physiology , Receptors, CCR7/metabolism , Receptors, CCR7/genetics , Encephalitis, California/virology , Encephalitis, California/genetics , Encephalitis, California/metabolism , Encephalitis, California/immunology , Mice, Inbred C57BL , Inflammation/metabolism , Inflammation/virology , Female , Male
2.
Sci Rep ; 11(1): 16424, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34385513

ABSTRACT

The California Serogroup (CSG) of Orthobunyaviruses comprises several viruses capable of causing neuroinvasive disease in humans, including La Crosse (LACV), Snowshoe Hare (SSHV), Tahyna (TAHV), Jamestown Canyon (JCV), and Inkoo (INKV) viruses. Diagnosis of specific CSG viruses is complicated by the high degree of antibody cross-reactivity between them, with laboratory standards requiring a fourfold higher titer of neutralizating antibody (NAb) activity to positively identify the etiologic virus. To help elucidate NAb relationships between neuroinvasive CSG viruses, we directly compared the cross-reactivity of NAb between LACV, SSHV, TAHV, JCV, and INKV. Mice were inoculated with individual viruses and the NAb activity of plasma samples was compared by plaque reduction neutralization tests against all five viruses. Overall, the results from these studies show that the CSG viruses induced high levels of NAb against the inoculum virus, and differing amounts of cross-reactive NAb against heterologous viruses. LACV, SSHV, and INKV elicited the highest amount of cross-reactive NAb. Interestingly, a fourfold difference in NAb titer between the inoculum virus and the other CSG viruses was not always observed. Thus, NAb titers, which are the gold-standard for diagnosing the etiologic agent for viral encephalitis, may not clearly differentiate between different CSG viruses.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cross Reactions , Encephalitis Virus, California/immunology , Encephalitis, California/immunology , Amino Acid Sequence , Animals , Encephalitis Virus, California/genetics , Female , Male , Mice , Sequence Homology, Amino Acid
3.
Virus Res ; 292: 198228, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33188797

ABSTRACT

Given the dual life cycle of arboviruses in insect and animal hosts and the importance of serum factors as a first line antiviral defense, we have examined the outcome of interactions between the arbovirus La Crosse Virus (LACV) and human serum. To mimic the life cycle between species, we used LACV derived from insect (I-LACV) and human keratinocyte (HaCaT) cells. Incubation of I-LACV with normal human serum did not result in neutralization, but instead stabilized I-LACV virions and enhanced the amount of infectious virus. Enhanced infectivity was also seen with heat-inactivated serum devoid of complement activity and with serum from a range of animals including mouse, ferret, and non-human primates. Depletion of antibodies from serum resulted in loss of enhancement of infectivity and sucrose gradient sedimentation assays showed IgG co-sedimenting with I-LACV particles. In agreement with our results with I-LACV, HaCaT-derived LACV was not neutralized by complement or antibodies in normal human serum. However, in contrast to I-LACV, HaCaT-derived LACV infectivity was stable when incubated alone and treatment with serum did not enhance infectivity. Our results indicate that LACV derived from insect cells differs substantially from virus derived from human cells, with I-LACV being dependent on serum factors to enhance infectivity. These findings suggest that understanding differential composition of insect versus animal cell-derived LACV may form the foundation for potential new antiviral approaches.


Subject(s)
Encephalitis, California/virology , Insecta/virology , Keratinocytes/virology , La Crosse virus/physiology , Serum/immunology , Animals , Cell Line , Disease Models, Animal , Encephalitis, California/immunology , Ferrets , Host-Pathogen Interactions , Humans , Keratinocytes/immunology , La Crosse virus/genetics , La Crosse virus/immunology , Mice , Neutralization Tests , Primates , Virus Replication
4.
J Neuroinflammation ; 16(1): 229, 2019 Nov 18.
Article in English | MEDLINE | ID: mdl-31739796

ABSTRACT

BACKGROUND: La Crosse virus (LACV) is the leading cause of pediatric arboviral encephalitis in the USA. LACV encephalitis can result in learning and memory deficits, which may be due to infection and apoptosis of neurons in the brain. Despite neurons being the primary cell infected in the brain by LACV, little is known about neuronal responses to infection. METHODS: Human cerebral organoids (COs), which contain a spectrum of developing neurons, were used to examine neuronal responses to LACV. Plaque assay and quantitative reverse transcription (qRT) PCR were used to determine the susceptibility of COs to LACV infection. Immunohistochemistry, flow cytometry, and single-cell transcriptomics were used to determine specific neuronal subpopulation responses to the virus. RESULTS: Overall, LACV readily infected COs causing reduced cell viability and increased apoptosis. However, it was determined that neurons at different stages of development had distinct responses to LACV. Both neural progenitors and committed neurons were infected with LACV, however, committed neurons underwent apoptosis at a higher rate. Transcriptomic analysis showed that committed neurons expressed fewer interferon (IFN)-stimulated genes (ISGs) and genes involved IFN signaling in response to infection compared to neural progenitors. Furthermore, induction of interferon signaling in LACV-infected COs by application of recombinant IFN enhanced cell viability. CONCLUSIONS: These findings indicate that neuronal maturation increases the susceptibility of neurons to LACV-induced apoptosis. This susceptibility is likely due, at least in part, to mature neurons being less responsive to virus-induced IFN as evidenced by their poor ISG response to LACV. Furthermore, exogenous administration of recombinant IFN to LACV COs rescued cellular viability suggesting that increased IFN signaling is overall protective in this complex neural tissue. Together these findings indicate that induction of IFN signaling in developing neurons is an important deciding factor in virus-induced cell death.


Subject(s)
Encephalitis, California/immunology , Interferon Type I/immunology , Neural Stem Cells/virology , Neurons/virology , Apoptosis/physiology , Cells, Cultured , Encephalitis, California/pathology , Humans , Induced Pluripotent Stem Cells , Neural Stem Cells/pathology , Neurons/cytology , Neurons/pathology , Organoids
5.
Viruses ; 11(3)2019 03 26.
Article in English | MEDLINE | ID: mdl-30917612

ABSTRACT

La Crosse virus (LACV) is a mosquito-transmitted arbovirus and the main cause of virus-mediated neurological diseases in children. To date, little is known about the role of C-type lectin receptors (CLRs)-an important class of pattern recognition receptors-in LACV recognition. DC-SIGN remains the only well-described CLR that recognizes LACV. In this study, we investigated the role of additional CLR/LACV interactions. To this end, we applied a flow-through chromatography method for the purification of LACV to perform an unbiased high-throughput screening of LACV with a CLR-hFc fusion protein library. Interestingly, the CARD9-associated CLRs Mincle, Dectin-1, and Dectin-2 were identified to strongly interact with LACV. Since CARD9 is a common adaptor protein for signaling via Mincle, Dectin-1, and Dectin-2, we performed LACV infection of Mincle-/- and CARD9-/- DCs. Mincle-/- and CARD9-/- DCs produced less amounts of proinflammatory cytokines, namely IL-6 and TNF-α, albeit no reduction of the LACV titer was observed. Together, novel CLR/LACV interactions were identified; however, the Mincle/CARD9 axis plays a limited role in early antiviral responses against LACV.


Subject(s)
CARD Signaling Adaptor Proteins/immunology , Encephalitis, California/immunology , Immunity, Innate , Lectins, C-Type/immunology , Membrane Proteins/immunology , Signal Transduction/immunology , Animals , Antiviral Agents , CARD Signaling Adaptor Proteins/genetics , Cytokines/immunology , Dendritic Cells/immunology , La Crosse virus/immunology , Lectins, C-Type/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Th17 Cells/immunology
6.
J Immunol ; 200(2): 471-476, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29246952

ABSTRACT

Inflammatory monocyte (iMO) recruitment to the brain is a hallmark of many neurologic diseases. Prior to entering the brain, iMOs must egress into the blood from the bone marrow through a mechanism, which for known encephalitic viruses, is CCR2 dependent. In this article, we show that during La Crosse Virus-induced encephalitis, egress of iMOs was surprisingly independent of CCR2, with similar percentages of iMOs in the blood and brain of heterozygous and CCR2-/- mice following infection. Interestingly, CCR2 was required for iMO trafficking from perivascular areas to sites of virus infection within the brain. Thus, CCR2 was not essential for iMO trafficking to the blood or the brain but was essential for trafficking within the brain parenchyma. Analysis of other orthobunyaviruses showed that Jamestown Canyon virus also induced CCR2-independent iMO egress to the blood. These studies demonstrate that the CCR2 requirement for iMO egress to the blood is not universal for all viruses.


Subject(s)
Antigens, Ly/metabolism , Encephalitis, California/immunology , Encephalitis, California/metabolism , La Crosse virus , Monocytes/immunology , Monocytes/metabolism , Receptors, CCR2/metabolism , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Brain/immunology , Brain/metabolism , Brain/pathology , Brain/virology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Encephalitis, California/virology , Female , Male , Mice , Mice, Transgenic , Monocytes/pathology
7.
J Neuroinflammation ; 14(1): 62, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28340587

ABSTRACT

BACKGROUND: La Crosse Virus (LACV) is a primary cause of pediatric viral encephalitis in the USA and can result in severe clinical outcomes. Almost all cases of LACV encephalitis occur in children 16 years or younger, indicating an age-related susceptibility. This susceptibility is recapitulated in a mouse model where weanling (3 weeks old or younger) mice are susceptible to LACV-induced disease, and adults (greater than 6 weeks) are resistant. Disease in mice and humans is associated with infiltrating leukocytes to the CNS. However, what cell types are infiltrating into the brain during virus infection and how these cells influence pathogenesis remain unknown. METHODS: In the current study, we analyzed lymphocytes recruited to the CNS during LACV-infection in clinical mice, using flow cytometry. We analyzed the contribution of these lymphocytes to LACV pathogenesis in weanling mice using knockout mice or antibody depletion. Additionally, we studied at the potential role of these lymphocytes in preventing LACV neurological disease in resistant adult mice. RESULTS: In susceptible weanling mice, disease was associated with infiltrating lymphocytes in the CNS, including NK cells, CD4 T cells, and CD8 T cells. Surprisingly, depletion of these cells did not impact neurological disease, suggesting these cells do not contribute to virus-mediated damage. In contrast, in disease-resistant adult animals, depletion of both CD4 T cells and CD8 T cells or depletion of B cells increased neurological disease, with higher levels of virus in the brain. CONCLUSIONS: Our current results indicate that lymphocytes do not influence neurological disease in young mice, but they have a critical role protecting adult animals from LACV pathogenesis. Although LACV is an acute virus infection, these studies indicate that the innate immune response in adults is not sufficient for protection and that components of the adaptive immune response are necessary to prevent virus from invading the CNS.


Subject(s)
Encephalitis, California/immunology , Lymphocytes/immunology , Animals , Disease Models, Animal , La Crosse virus , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
J Virol ; 88(19): 11070-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25008929

ABSTRACT

UNLABELLED: La Crosse virus (LACV) is the major cause of pediatric viral encephalitis in the United States; however, the mechanisms responsible for age-related susceptibility in the pediatric population are not well understood. Our current studies in a mouse model of LACV infection indicated that differences in myeloid dendritic cell (mDC) responses between weanling and adult mice accounted for susceptibility to LACV-induced neurological disease. We found that type I interferon (IFN) responses were significantly stronger in adult than in weanling mice. Production of these IFNs required both endosomal Toll-like receptors (TLRs) and cytoplasmic RIG-I-like receptors (RLRs). Surprisingly, IFN expression was not dependent on plasmacytoid DCs (pDCs) but rather was dependent on mDCs, which were found in greater number and induced stronger IFN responses in adults than in weanlings. Inhibition of these IFN responses in adults resulted in susceptibility to LACV-induced neurological disease, whereas postinfection treatment with type I IFN provided protection in young mice. These studies provide a definitive mechanism for age-related susceptibility to LACV encephalitis, where mDCs in young mice are insufficiently activated to control peripheral virus replication, thereby allowing virus to persist and eventually cause central nervous system (CNS) disease. IMPORTANCE: La Crosse virus (LACV) is the primary cause of pediatric viral encephalitis in the United States. Although the virus infects both adults and children, over 80% of the reported neurological disease cases are in children. To understand why LACV causes neurological disease primarily in young animals, we used a mouse model where weanling mice, but not adult mice, develop neurological disease following virus infection. We found that an early immune response cell type, myeloid dendritic cells, was critical for protection in adult animals and that these cells were reduced in young animals. Activation of these cells during virus infection or after treatment with type I interferon in young animals provided protection from LACV. Thus, this study demonstrates a reason for susceptibility to LACV infection in young animals and shows that early therapeutic treatment in young animals can prevent neurological disease.


Subject(s)
Central Nervous System/immunology , Dendritic Cells/immunology , Encephalitis, California/immunology , La Crosse virus/immunology , Myeloid Cells/immunology , Age Factors , Animals , Animals, Newborn , Central Nervous System/virology , Dendritic Cells/virology , Disease Models, Animal , Disease Susceptibility , Encephalitis, California/mortality , Encephalitis, California/virology , Gene Expression/immunology , Humans , Injections, Intradermal , Injections, Intraperitoneal , Injections, Intraventricular , Interferon Type I/genetics , Interferon Type I/immunology , Mice , Myeloid Cells/virology , Survival Analysis , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Virus Replication
9.
J Neurovirol ; 20(2): 150-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23846288

ABSTRACT

Viral encephalitis represents a significant, and costly, public health threat particularly for high-risk pediatric populations. An emerging mosquito-borne pathogen endemic to the United States, La Crosse virus (LACV) is one of the most common causes of viral encephalitis in children in the United States. However, no licensed therapeutics or vaccines currently exist for treatment. Hampering development efforts, the host response to LACV and its role in disease pathogenesis has only recently been examined. In this review, we discuss the current understanding of innate immune response in the context of viral pathogenesis and host susceptibility to LACV. In addition, we address the need for a clearer understanding of the early host-virus interactions in LACV infections as it relates to viral pathogenesis in the central nervous system.


Subject(s)
Central Nervous System/immunology , Disease Reservoirs/veterinary , Encephalitis, California/immunology , Immunity, Innate , La Crosse virus/immunology , Aedes/virology , Animals , Central Nervous System/pathology , Central Nervous System/virology , Child , Disease Reservoirs/virology , Disease Susceptibility , Disease Vectors , Encephalitis, California/pathology , Encephalitis, California/transmission , Encephalitis, California/virology , Host Specificity , Host-Pathogen Interactions , Humans , Interferon Type I/biosynthesis , Mice
10.
Immunity ; 38(4): 705-16, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-23499490

ABSTRACT

La Crosse virus (LACV), a zoonotic Bunyavirus, is a major cause of pediatric viral encephalitis in the United States. A hallmark of neurological diseases caused by LACV and other encephalitic viruses is the induction of neuronal cell death. Innate immune responses have been implicated in neuronal damage, but no mechanism has been elucidated. By using in vitro studies in primary neurons and in vivo studies in mice, we have shown that LACV infection induced the RNA helicase, RIG-I, and mitochondrial antiviral signaling protein (MAVS) signaling pathway, resulting in upregulation of the sterile alpha and TIR-containing motif 1 (SARM1), an adaptor molecule that we found to be directly involved in neuronal damage. SARM1-mediated cell death was associated with induced oxidative stress response and mitochondrial damage. These studies provide an innate-immune signaling mechanism for virus-induced neuronal death and reveal potential targets for development of therapeutics to treat encephalitic viral infections.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis , Armadillo Domain Proteins/metabolism , Cytoskeletal Proteins/metabolism , Encephalitis, California/immunology , La Crosse virus/immunology , Mitochondria/metabolism , Neurons/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Armadillo Domain Proteins/genetics , Cells, Cultured , Cytoskeletal Proteins/genetics , Encephalitis, California/complications , Encephalitis, California/drug therapy , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Neurons/virology , Oxidative Stress , Primary Cell Culture , Signal Transduction/immunology , Up-Regulation
11.
J Virol ; 86(20): 11223-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875966

ABSTRACT

Beta interferon (IFN-ß) is a major component of innate immunity in mammals, but information on the in vivo source of this cytokine after pathogen infection is still scarce. To identify the cell types responsible for IFN-ß production during viral encephalitis, we used reporter mice that express firefly luciferase under the control of the IFN-ß promoter and stained organ sections with luciferase-specific antibodies. Numerous luciferase-positive cells were detected in regions of La Crosse virus (LACV)-infected mouse brains that contained many infected cells. Double-staining experiments with cell-type-specific markers revealed that similar numbers of astrocytes and microglia of infected brains were luciferase positive, whereas virus-infected neurons rarely contained detectable levels of luciferase. Interestingly, if a mutant LACV unable of synthesizing the IFN-antagonistic factor NSs was used for challenge, the vast majority of the IFN-ß-producing cells in infected brains were astrocytes rather than microglia. Similar conclusions were reached in a second series of experiments in which conditional reporter mice expressing the luciferase reporter gene solely in defined cell types were infected with wild-type or mutant LACV. Collectively, our data suggest that glial cells rather than infected neurons represent the major source of IFN-ß in LACV-infected mouse brains. They further indicate that IFN-ß synthesis in astrocytes and microglia is differentially affected by the viral IFN antagonist, presumably due to differences in LACV susceptibility of these two cell types.


Subject(s)
Astrocytes/immunology , Encephalitis, California/immunology , Interferon-beta/biosynthesis , La Crosse virus/immunology , Microglia/immunology , Animals , Astrocytes/metabolism , Astrocytes/virology , Brain/metabolism , Brain/virology , Interferon-beta/genetics , Interferon-beta/immunology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Microglia/virology , Neurons/immunology , Neurons/metabolism , Neurons/virology , Promoter Regions, Genetic
12.
J Virol ; 86(1): 420-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22013033

ABSTRACT

La Crosse virus (LACV) and Jamestown Canyon virus (JCV), family Bunyaviridae, are mosquito-borne viruses that are endemic in North America and recognized as etiologic agents of encephalitis in humans. Both viruses belong to the California encephalitis virus serogroup, which causes 70 to 100 cases of encephalitis a year. As a first step in creating live attenuated viral vaccine candidates for this serogroup, we have generated a recombinant LACV expressing the attachment/fusion glycoproteins of JCV. The JCV/LACV chimeric virus contains full-length S and L segments derived from LACV. For the M segment, the open reading frame (ORF) of LACV is replaced with that derived from JCV and is flanked by the untranslated regions of LACV. The resulting chimeric virus retained the same robust growth kinetics in tissue culture as observed for either parent virus, and the virus remains highly infectious and immunogenic in mice. Although both LACV and JCV are highly neurovirulent in 21 day-old mice, with 50% lethal dose (LD50) values of 0.1 and 0.5 log10 PFU, respectively, chimeric JCV/LACV is highly attenuated and does not cause disease even after intracerebral inoculation of 10³ PFU. Parenteral vaccination of mice with 10¹ or 10³ PFU of JCV/LACV protected against lethal challenge with LACV, JCV, and Tahyna virus (TAHV). The chimeric virus was infectious and immunogenic in rhesus monkeys and induced neutralizing antibodies to JCV, LACV, and TAHV. When vaccinated monkeys were challenged with JCV, they were protected against the development of viremia. Generation of highly attenuated yet immunogenic chimeric bunyaviruses could be an efficient general method for development of vaccines effective against these pathogenic viruses.


Subject(s)
Chimera/immunology , Encephalitis Virus, California/genetics , Encephalitis, California/prevention & control , Gene Expression , La Crosse virus/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Viral/immunology , Cell Line , Chimera/genetics , Chimera/growth & development , Encephalitis, California/immunology , Encephalitis, California/virology , Humans , La Crosse virus/genetics , La Crosse virus/growth & development , Macaca mulatta , Mice , Vaccination , Viral Envelope Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology
13.
Zoonoses Public Health ; 59(3): 181-3, 2012 May.
Article in English | MEDLINE | ID: mdl-21848528

ABSTRACT

We evaluated the validity of single versus paired serologic testing for La Crosse virus (LACV) encephalitis surveillance. Compared with paired serology, a single positive IgG or IgM immunoflourescent antibody titre appears useful for LACV encephalitis surveillance with sensitivity, 75%; specificity, 98%; positive predictive value, 95%; and overall test efficiency 92%.


Subject(s)
Encephalitis, California/diagnosis , Immunoglobulin G/analysis , Immunoglobulin M/analysis , La Crosse virus/immunology , Serologic Tests/methods , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Viral/analysis , Antibodies, Viral/immunology , Antigens, Viral/analysis , Antigens, Viral/immunology , Child , Child, Preschool , Encephalitis, California/immunology , Encephalitis, California/virology , Female , Fluorescent Antibody Technique , Hospitalization/statistics & numerical data , Humans , Infant , La Crosse virus/isolation & purification , Male , Middle Aged , Predictive Value of Tests , Reproducibility of Results , Sentinel Surveillance , Tennessee/epidemiology , Young Adult
14.
Virol J ; 8: 135, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21435229

ABSTRACT

BACKGROUND: Tahyna virus (TAHV) is a human pathogen of the California encephalitis virus (CEV) serogroup (Bunyaviridae) endemic to Europe, Asia, and Africa. TAHV maintains an enzootic life cycle with several species of mosquito vectors and hares, rabbits, hedgehogs, and rodents serving as small mammal amplifying hosts. Human TAHV infection occurs in summer and early fall with symptoms of fever, headache, malaise, conjunctivitis, pharyngitis, and nausea. TAHV disease can progress to CNS involvement, although unlike related La Crosse virus (LACV), fatalities have not been reported. Human infections are frequent with neutralizing antibodies present in 60-80% of the elderly population in endemic areas. RESULTS: In order to determine the genomic sequence of wild-type TAHV, we chose three TAHV isolates collected over a 26-year period from mosquitoes. Here we present the first complete sequence of the TAHV S, M, and L segments. The three TAHV isolates maintained a highly conserved genome with both nucleotide and amino acid sequence identity greater than 99%. In order to determine the extent of genetic relatedness to other members of the CEV serogroup, we compared protein sequences of TAHV with LACV, Snowshoe Hare virus (SSHV), Jamestown Canyon virus (JCV), and Inkoo virus (INKV). By amino acid comparison, TAHV was most similar to SSHV followed by LACV, JCV, and INKV. The sequence of the GN protein is most conserved followed by L, N, GC, NSS, and NSM. In a weanling Swiss Webster mouse model, all three TAHV isolates were uniformly neurovirulent, but only one virus was neuroinvasive. In rhesus monkeys, the virus was highly immunogenic even in the absence of viremia. Cross neutralization studies utilizing monkey immune serum demonstrated that TAHV is antigenically distinct from North American viruses LACV and JCV. CONCLUSIONS: Here we report the first complete sequence of TAHV and present genetic analysis of new-world viruses, LACV, SSHV, and JCV with old-world viruses, TAHV and INKV. Using immune serum generated in monkeys against TAHV, LACV, and JCV, we have demonstrated cross-neutralization within the CEV serogroup. Such cross reactivity may complicate virus identification, especially following JCV infection which elicited antibodies that cross neutralized both LACV and TAHV. These data also suggest that a single vaccine could generate a cross-neutralizing antibody response which may provide protection against CEV serogroup viruses from a wide geographic range.


Subject(s)
Disease Models, Animal , Encephalitis Virus, California/genetics , Encephalitis Virus, California/pathogenicity , Encephalitis, California/immunology , Macaca mulatta , Mice , Animals , Antibodies, Viral/immunology , Base Sequence , Culicidae/virology , Encephalitis Virus, California/classification , Encephalitis Virus, California/immunology , Encephalitis, California/virology , Humans , Molecular Sequence Data , Rabbits , Virulence
15.
J Virol ; 73(8): 6984-91, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10400797

ABSTRACT

The human MxA protein is part of the antiviral state induced by alpha/beta interferon (IFN-alpha/beta). MxA inhibits the multiplication of several RNA viruses in cell culture. However, its antiviral potential in vivo has not yet been fully explored. We have generated MxA-transgenic mice that lack a functional IFN system by crossing MxA-transgenic mice constitutively expressing MxA with genetically targeted (knockout) mice lacking the beta subunit of the IFN-alpha/beta receptor (IFNAR-1(-/-) mice). These mice are an ideal animal model to investigate the unique antiviral activity of human MxA in vivo, because they are unable to express other IFN-induced proteins. Here, we show that MxA confers resistance to Thogoto virus, La Crosse virus, and Semliki Forest virus. No Thogoto virus progeny was detectable in MxA-transgenic mice, indicating an efficient block of virus replication at the primary site of infection. In the case of La Crosse virus, MxA restricted invasion of the central nervous system. In contrast, Semliki Forest virus multiplication in the brain was detectable in both MxA-expressing and nonexpressing IFNAR-1(-/-) mice. However, viral titers were clearly reduced in MxA-transgenic mice. Our results demonstrate that MxA does not need the help of other IFN-induced proteins for activity but is a powerful antiviral agent on its own. Moreover, the results suggest that MxA may protect humans from potential fatal infections by La Crosse virus and other viral pathogens.


Subject(s)
Antiviral Agents/immunology , Encephalitis, California/immunology , GTP-Binding Proteins , Interferon-alpha/immunology , Interferon-beta/immunology , La Crosse virus/immunology , Proteins/immunology , Receptors, Interferon/immunology , Alphavirus Infections/immunology , Animals , Antiviral Agents/biosynthesis , Antiviral Agents/genetics , Humans , La Crosse virus/growth & development , Membrane Proteins , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Myxovirus Resistance Proteins , Orthomyxoviridae Infections/immunology , Protein Biosynthesis , Proteins/genetics , Receptor, Interferon alpha-beta , Receptors, Interferon/genetics , Semliki forest virus/growth & development , Thogotovirus/immunology
16.
Hum Gene Ther ; 10(10): 1649-58, 1999 Jul 01.
Article in English | MEDLINE | ID: mdl-10428210

ABSTRACT

La Crosse virus (LACV)-mediated encephalitis is the most frequently reported arboviral disease in the United States, but to date no vaccine against this virus is available. We have established a new animal model, genetically targeted mice lacking a functional interferon type I receptor (IFNAR-1). These mice show an age-independent susceptibility to LACV and develop an acute encephalitis within 6 days of infection, thereby allowing the evaluation of vaccines against LACV. Taking advantage of this knockout mouse model, we have assessed the feasibility of DNA vaccination against this viral disease. Plasmid DNAs, encoding either the virus surface glycoproteins G1 and G2 or the internal nucleocapsid protein N, were used to immunize IFNAR-1-deficient mice. Mice vaccinated with DNA encoding the glycoproteins G1 and G2 produced neutralizing antibodies and exhibited a high degree of protection against challenge with high doses of LACV. Depletion of CD4+ T cells in mice vaccinated with DNA encoding G1/G2 reduced their capacity to control the infection. Virus titration and immunohistological analysis revealed that the protected mice showed no evidence of LACV particles in the brain. This indicates that the vaccine-induced immune response efficiently blocked viral spreading from the primary replication site to the brain. In contrast, immunization with DNA encoding protein N yielded only a partial protective effect that can be attributed to the cellular immune response. Taken together, this study shows that DNA vaccines can be designed to efficiently induce a protective immune response based on neutralizing antibodies and CD4+ T cells.


Subject(s)
Antibodies, Viral/immunology , CD4-Positive T-Lymphocytes/immunology , Encephalitis, California/prevention & control , La Crosse virus/immunology , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Cell Line , Cell Line, Transformed , Chlorocebus aethiops , Cricetinae , DNA, Viral/immunology , Disease Models, Animal , Encephalitis, California/immunology , Gene Expression , Glycoproteins/genetics , Glycoproteins/immunology , Mice , Mice, Knockout , Neutralization Tests , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/immunology , Vaccination , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
17.
Am J Trop Med Hyg ; 54(4): 332-7, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8615442

ABSTRACT

On the Cherokee Indian Reservation and surrounding area of western North Carolina, an area-wide serosurvey was conducted to determine the prevalence of neutralizing antibody to La Crosse (LAC) virus. A questionnaire was used to identify risk factors important in exposure to virus-infected mosquitoes in populations near the reservation. Of 1,008 serum samples tested, 9.6% were positive for LAC virus antibody. For samples solely collected from on (n = 311) or off (n = 697) the reservation, the prevalence of seropositive samples was 20.6% on the reservation and only 4.7% off the reservation. Seropositivity increased directly with age, indicating that transmission of LAC virus was highly endemic. Age and location residence (on versus off the reservation) were significant risk factors for exposure to LAC virus. Persons on the reservation were 5.5 times more likely to have been exposed to LAC virus than were people who reside off the reservation. An additive increase in risk of 1.5 times over each age group was found, so that the oldest age group ( > or = 75 years) was 7.5 times more likely to have been exposed to LAC virus than was the youngest age group ( < 1-14 years).


Subject(s)
Antibodies, Viral/blood , Encephalitis, California/epidemiology , Indians, North American , La Crosse virus/immunology , Adolescent , Adult , Age Distribution , Aged , Animals , Child , Child, Preschool , Chlorocebus aethiops , Encephalitis, California/immunology , Female , Humans , Incidence , Infant , Male , Middle Aged , Neutralization Tests , North Carolina/epidemiology , Risk Factors , Seasons , Seroepidemiologic Studies , United States/epidemiology , Vero Cells
18.
Am J Trop Med Hyg ; 54(4): 338-42, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8615443

ABSTRACT

To further understand the role of wild mammals in the maintenance of La Crosse virus (LACV) in nature, we investigated the effects of inoculation method and virus source on the duration and amplitude of LACV viremia in vertebrate hosts. Earlier work suggested that deer are not sufficiently susceptible to LACV to play an important role in its maintenance. We re-evaluated the susceptibility of deer since subsequent studies showed that they constitute 65% of Aedes triseriatus blood meals, and thus would be exposed frequently to the virus. In our study, deer developed higher and longer viremia following exposure to LACV by infected Ae. triseriatus than those previously reported by inoculation with needle and syringe. However, susceptible Ae. triseriatus that fed on these viremic animals did not become infected. Because a large number of uninfected mosquitoes can feed upon a viremic deer in nature, we believe that deer should not be disregarded completely as a possible amplifier in the LACV transmission cycle. We also infected chipmunks to determine if there were significant differences in viremia response from mosquito delivery of virus to the chipmunk host, compared with artificial exposure by injection. Chipmunks exposed to infected mosquitoes had higher and longer viremias than the ones produced by intramuscular injection of an LACV suspension. These findings show the importance of using LACV infected mosquitoes for transmission experiments in mammals.


Subject(s)
Deer , Encephalitis, California/veterinary , La Crosse virus/isolation & purification , Rodent Diseases/transmission , Sciuridae , Viremia/veterinary , Aedes/virology , Animals , Animals, Suckling , Antibodies, Viral/blood , Disease Reservoirs , Disease Susceptibility , Encephalitis, California/immunology , Encephalitis, California/transmission , Female , Insect Vectors/virology , La Crosse virus/immunology , Mice , Rodent Diseases/immunology , Viremia/immunology , Viremia/transmission
19.
J Virol ; 69(6): 3475-81, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7745694

ABSTRACT

La Crosse virus, a member of the California serogroup of bunyaviruses, is an important cause of pediatric encephalitis in the midwestern United States. Like all bunyaviruses, La Crosse virus contains two glycoproteins, G1 and G2, the larger of which, G1, is the target of neutralizing antibodies. To develop an understanding of the role of each of the glycoproteins in the generation of a protective immune response, we immunized 1-week-old mice with three different preparations: a vaccinia virus recombinant (VV.ORF) that expresses both G1 and G2, a vaccinia virus recombinant (VV.G1) that expresses G1 only, and a truncated soluble G1 (sG1) protein prepared in a baculovirus system. Whereas VV.ORF generated a protective response that was mostly directed against G1, VV.G1 was only partially effective at inducing a neutralizing response and at protecting mice from a potentially lethal challenge with La Crosse virus. Nevertheless, a single immunization with the sG1 preparation resulted in a robust immune response and protection against La Crosse virus. These results indicate that (i) the G1 protein by itself can induce an immune response sufficient for protection from a lethal challenge with La Crosse virus, (ii) a neutralizing humoral response correlates with protection, and (iii) the context in which G1 is presented affects its immunogenicity. The key step in the defense against central nervous system infection appeared to be interruption of a transient viremia that occurred just after La Crosse virus inoculation.


Subject(s)
Antibodies, Viral/immunology , Encephalitis, California/prevention & control , Glycoproteins/immunology , La Crosse virus/immunology , Viral Proteins/immunology , Aging/immunology , Animals , Cattle , Cell Line , Disease Susceptibility/immunology , Encephalitis, California/immunology , Glycoproteins/genetics , Glycoproteins/therapeutic use , Immunotherapy, Adoptive , La Crosse virus/metabolism , La Crosse virus/pathogenicity , Mice , Neutralization Tests , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use , Vaccines, Synthetic/immunology , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/therapeutic use , Viral Vaccines/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...