Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(3): e0262967, 2022.
Article in English | MEDLINE | ID: mdl-35298486

ABSTRACT

Herein we describe a previously uninvestigated salivary gland escape barrier (SEB) in Culex tarsalis mosquitoes infected with two different strains of Western equine encephalitis virus (WEEV). The WEEV strains were originally isolated either from mosquitoes (IMP181) or a human patient (McMillan). Both IMP181 and McMillan viruses were fully able to infect the salivary glands of Culex tarsalis after intrathoracic injection as determined by expression of mCherry fluorescent protein. IMP181, however, was better adapted to transmission as measured by virus titer in saliva as well as transmission rates in infected mosquitoes. We used chimeric recombinant WEEV strains to show that inclusion of IMP181-derived structural genes partially circumvents the SEB.


Subject(s)
Culex , Encephalomyelitis, Equine , Animals , Disease Vectors , Encephalitis Virus, Western Equine/genetics , Humans , Mosquito Vectors , Salivary Glands
2.
Exp Neurol ; 346: 113845, 2021 12.
Article in English | MEDLINE | ID: mdl-34454938

ABSTRACT

Viral infection of the central nervous system (CNS) can cause lasting neurological decline in surviving patients and can present with symptoms resembling Parkinson's disease (PD). The mechanisms underlying postencephalitic parkinsonism remain unclear but are thought to involve increased innate inflammatory signaling in glial cells, resulting in persistent neuroinflammation. We therefore studied the role of glial cells in regulating neuropathology in postencephalitic parkinsonism by studying the involvement of astrocytes in loss of dopaminergic neurons and aggregation of α-synuclein protein following infection with western equine encephalitis virus (WEEV). Infections were conducted in both wildtype mice and in transgenic mice lacking NFκB inflammatory signaling in astrocytes. For 2 months following WEEV infection, we analyzed glial activation, neuronal loss and protein aggregation across multiple brain regions, including the substantia nigra pars compacta (SNpc). These data revealed that WEEV induces loss of SNpc dopaminergic neurons, persistent activation of microglia and astrocytes that precipitates widespread aggregation of α-synuclein in the brain of C57BL/6 mice. Microgliosis and macrophage infiltration occurred prior to activation of astrocytes and was followed by opsonization of ⍺-synuclein protein aggregates in the cortex, hippocampus and midbrain by the complement protein, C3. Astrocyte-specific NFκB knockout mice had reduced gliosis, α-synuclein aggregate formation and neuronal loss. These data suggest that astrocytes play a critical role in initiating PD-like pathology following encephalitic infection with WEEV through innate immune inflammatory pathways that damage dopaminergic neurons, possibly by hindering clearance of ⍺-synuclein aggregates. Inhibiting glial inflammatory responses could therefore represent a potential therapy strategy for viral parkinsonism.


Subject(s)
Astrocytes/metabolism , Dopaminergic Neurons/metabolism , Encephalitis, Viral/metabolism , Inflammation Mediators/metabolism , Protein Aggregates/physiology , alpha-Synuclein/metabolism , Animals , Astrocytes/immunology , Dopaminergic Neurons/immunology , Encephalitis Virus, Western Equine/immunology , Encephalitis Virus, Western Equine/metabolism , Encephalitis, Viral/immunology , Female , Humans , Inflammation Mediators/immunology , Male , Mice , Mice, Knockout , Signal Transduction/physiology
3.
J Virol ; 94(3)2020 01 17.
Article in English | MEDLINE | ID: mdl-31694937

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is one of the important human and animal pathogens. It forms replication enzyme complexes (RCs) containing viral nonstructural proteins (nsPs) that mediate the synthesis of virus-specific RNAs. The assembly and associated functions of RC also depend on the presence of a specific set of host proteins. Our study demonstrates that the hypervariable domain (HVD) of VEEV nsP3 interacts with the members of the FXR family of cellular proteins and also binds the Src homology 3 (SH3) domain-containing proteins CD2AP and SH3KBP1. Interactions with FXR family members are mediated by the C-terminal repeating peptide of HVD. A single short, minimal motif identified in this study is sufficient for driving efficient VEEV replication in the absence of HVD interactions with other host proteins. The SH3 domain-containing proteins bind to another fragment of VEEV HVD. They can promote viral replication in the absence of FXR-HVD interactions albeit less efficiently. VEEV replication can be also switched from an FXR-dependent to a chikungunya virus-specific, G3BP-dependent mode. The described modifications of VEEV HVD have a strong impact on viral replication in vitro and pathogenesis. Their effects on viral pathogenesis depend on mouse age and the genetic background of the virus.IMPORTANCE The replication of alphaviruses is determined by specific sets of cellular proteins, which mediate the assembly of viral replication complexes. Some of these critical host factors interact with the hypervariable domain (HVD) of alphavirus nsP3. In this study, we have explored binding sites of host proteins, which are specific partners of nsP3 HVD of Venezuelan equine encephalitis virus. We also define the roles of these interactions in viral replication both in vitro and in vivo A mechanistic understanding of the binding of CD2AP, SH3KBP1, and FXR protein family members to VEEV HVD uncovers important aspects of alphavirus evolution and determines new targets for the development of alphavirus-specific drugs and directions for viral attenuation and vaccine development.


Subject(s)
Encephalitis Virus, Venezuelan Equine/genetics , Mutation , Protein Interaction Domains and Motifs , Viral Nonstructural Proteins/genetics , Virus Replication/genetics , Adaptor Proteins, Signal Transducing , Animals , Binding Sites , Cell Line , Chikungunya virus/metabolism , Cytoskeletal Proteins , Disease Models, Animal , Encephalomyelitis, Venezuelan Equine/virology , Humans , Intrinsically Disordered Proteins/metabolism , Mice , Sequence Alignment , Viral Nonstructural Proteins/chemistry , src Homology Domains
4.
NPJ Parkinsons Dis ; 5: 20, 2019.
Article in English | MEDLINE | ID: mdl-31531390

ABSTRACT

Neuroinvasive infections with mosquito-borne alphaviruses such as Western equine encephalitis virus (WEEV) can cause post-encephalitic parkinsonism. To understand the mechanisms underlying these neurological effects, we examined the capacity of WEEV to induce progressive neurodegeneration in outbred CD-1 mice following non-lethal encephalitic infection. Animals were experientally infected with recombinant WEEV expressing firefly luciferase or dsRed (RFP) reporters and the extent of viral replication was controlled using passive immunotherapy. WEEV spread along the neuronal axis from the olfactory bulb to the entorhinal cortex, hippocampus and basal midbrain by 4 days post infection (DPI). Infection caused activation of microglia and astrocytes, selective loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neurobehavioral abnormalities. After 8 weeks, surviving mice displayed continued loss of dopamine neurons in the SNpc, lingering glial cell activation and gene expression profiles consistent with a neurodegenerative phenotype. Strikingly, prominent proteinase K-resistant protein aggregates were present in the the entorhinal cortex, hippocampus and basal midbrain that stained positively for phospho-serine129 α-synuclein (SNCA). These results indicate that WEEV may cause lasting neurological deficits through a severe neuroinflammatory response promoting both neuronal injury and protein aggregation in surviving individuals.

5.
Virology ; 534: 14-24, 2019 08.
Article in English | MEDLINE | ID: mdl-31163352

ABSTRACT

Chikungunya virus (CHIKV) is an important arthritogenic human pathogen that is already circulating in both hemispheres. In the present study, we substituted VLoop, located on the surface of nsP2, by other amino acid sequences. These modifications had deleterious effects on viral nuclear functions and made CHIKV incapable of interfering with the induction of type I interferon and the antiviral response in both mouse and human cells. Importantly, the identified mutations have no significant effects on the synthesis of virus-specific RNAs and viral structural proteins. The designed mutants induced a few orders of magnitude lower viremia but remained highly immunogenic in mice. Thus, the proposed modifications of nsP2 can additionally improve the safety of the attenuated strain CHIKV 181/25. Furthermore, defined mutations in the macro domain of another nonstructural protein, nsP3, additionally reduce cytopathogenicity of nsP2 mutants in human cells, and can be potentially applied for CHIKV attenuation.


Subject(s)
Cell Nucleus/virology , Chikungunya Fever/virology , Chikungunya virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Chikungunya Fever/genetics , Chikungunya Fever/metabolism , Chikungunya virus/chemistry , Chikungunya virus/genetics , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mutation , Protein Domains , RNA, Viral/genetics , RNA, Viral/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
6.
Viruses ; 10(5)2018 05 02.
Article in English | MEDLINE | ID: mdl-29724035

ABSTRACT

New World arenaviruses cause fatal hemorrhagic disease in South America. Pirital virus (PIRV), a mammarenavirus hosted by Alston’s cotton rat (Sigmodon alstoni), causes a disease in Syrian golden hamsters (Mesocricetus auratus) (biosafety level-3, BSL-3) that has many pathologic similarities to the South American hemorrhagic fevers (BSL-4) and, thus, is considered among the best small-animal models for human arenavirus disease. Here, we extend in greater detail previously described clinical and pathological findings in Syrian hamsters and provide evidence for a pro-inflammatory macrophage response during PIRV infection. The liver was the principal target organ of the disease, and signs of Kupffer cell involvement were identified in mortally infected hamster histopathology data. Differential expression analysis of liver mRNA revealed signatures of the pro-inflammatory response, hematologic dysregulation, interferon pathway and other host response pathways, including 17 key transcripts that were also reported in two non-human primate (NHP) arenavirus liver-infection models, representing both Old and New World mammarenavirus infections. Although antigen presentation may differ among rodent and NHP species, key hemostatic and innate immune-response components showed expression parallels. Signatures of pro-inflammatory macrophage involvement in PIRV-infected livers included enrichment of Ifng, Nfkb2, Stat1, Irf1, Klf6, Il1b, Cxcl10, and Cxcl11 transcripts. Together, these data indicate that pro-inflammatory macrophage M1 responses likely contribute to the pathogenesis of acute PIRV infection.


Subject(s)
Arenaviridae Infections/immunology , Arenaviruses, New World/pathogenicity , Liver/immunology , Macrophages/immunology , Animals , Cricetinae , Disease Models, Animal , Gene Expression Profiling , Host-Pathogen Interactions , Immunity, Innate , Kupffer Cells/virology , Liver/pathology , Liver/virology
7.
Virology ; 499: 30-39, 2016 12.
Article in English | MEDLINE | ID: mdl-27632563

ABSTRACT

Eastern, Venezuelan and western equine encephalitis viruses (EEEV, VEEV, and WEEV) are mosquito-borne viruses that cause substantial disease in humans and other vertebrates. Vaccines are limited and current treatment options have not proven successful. In this report, we vaccinated outbred mice with lipid-antigen-nucleic acid-complexes (LANACs) containing VEEV E1+WEEV E1 antigen and characterized protective efficacy against lethal EEEV, VEEV, and WEEV challenge. Vaccination resulted in complete protection against EEEV, VEEV, and WEEV in CD-1 mice. Measurements of bioluminescence and plaque reduction neutralization tests (PRNTs) indicate that LANAC VEEV E1+WEEV E1 vaccination is sterilizing against VEEV and WEEV challenge; whereas immunity to EEEV is not sterilizing. Passive transfer of rabbit VEEV E1+WEEV E1 immune serum to naive mice extended the mean time to death (MTD) of EEEV challenged mice and provided significant protection from lethal VEEV and WEEV challenge.


Subject(s)
Alphavirus/immunology , Antigens, Viral/immunology , Cross Reactions/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Western Equine/immunology , Viral Proteins/immunology , Alphavirus Infections/immunology , Alphavirus Infections/mortality , Alphavirus Infections/prevention & control , Alphavirus Infections/virology , Animals , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Antigens, Viral/genetics , Cell Line , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/pathogenicity , Female , Gene Expression , Genes, Reporter , Immunity, Humoral , Immunization , Liposomes , Mice , Nucleic Acids , Sequence Homology , Viral Proteins/administration & dosage , Viral Proteins/genetics , Virulence/genetics , Virus Replication
8.
J Virol ; 90(12): 5785-96, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27053560

ABSTRACT

UNLABELLED: Venezuelan and western equine encephalitis viruses (VEEV and WEEV; Alphavirus; Togaviridae) are mosquito-borne pathogens causing central nervous system (CNS) disease in humans and equids. Adult CD-1 mice also develop CNS disease after infection with VEEV and WEEV. Adult CD-1 mice infected by the intranasal (i.n.) route, showed that VEEV and WEEV enter the brain through olfactory sensory neurons (OSNs). In this study, we injected the mouse footpad with recombinant WEEV (McMillan) or VEEV (subtype IC strain 3908) expressing firefly luciferase (fLUC) to simulate mosquito infection and examined alphavirus entry in the CNS. Luciferase expression served as a marker of infection detected as bioluminescence (BLM) by in vivo and ex vivo imaging. BLM imaging detected WEEV and VEEV at 12 h postinoculation (hpi) at the injection site (footpad) and as early as 72 hpi in the brain. BLM from WEEV.McM-fLUC and VEEV.3908-fLUC injections was initially detected in the brain's circumventricular organs (CVOs). No BLM activity was detected in the olfactory neuroepithelium or OSNs. Mice were also injected in the footpad with WEEV.McM expressing DsRed (Discosoma sp.) and imaged by confocal fluorescence microscopy. DsRed imaging supported our BLM findings by detecting WEEV in the CVOs prior to spreading along the neuronal axis to other brain regions. Taken together, these findings support our hypothesis that peripherally injected alphaviruses enter the CNS by hematogenous seeding of the CVOs followed by centripetal spread along the neuronal axis. IMPORTANCE: VEEV and WEEV are mosquito-borne viruses causing sporadic epidemics in the Americas. Both viruses are associated with CNS disease in horses, humans, and mouse infection models. In this study, we injected VEEV or WEEV, engineered to express bioluminescent or fluorescent reporters (fLUC and DsRed, respectively), into the footpads of outbred CD-1 mice to simulate transmission by a mosquito. Reporter expression serves as detectable bioluminescent and fluorescent markers of VEEV and WEEV replication and infection. Bioluminescence imaging, histological examination, and confocal fluorescence microscopy were used to identify early entry sites of these alphaviruses in the CNS. We observed that specific areas of the brain (circumventricular organs [CVOs]) consistently showed the earliest signs of infection with VEEV and WEEV. Histological examination supported VEEV and WEEV entering the brain of mice at specific sites where the blood-brain barrier is naturally absent.


Subject(s)
Blood-Brain Barrier/virology , Brain/virology , Encephalitis Virus, Venezuelan Equine/physiology , Encephalitis Virus, Western Equine/physiology , Encephalomyelitis, Venezuelan Equine/virology , Virus Internalization , Adult , Animals , Blood-Brain Barrier/physiopathology , Brain/pathology , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/growth & development , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/growth & development , Humans , Luciferases , Luminescent Measurements , Mice , Olfactory Receptor Neurons/virology , Optical Imaging/methods , Viral Load
9.
Mol Pharmacol ; 87(6): 1021-34, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25858541

ABSTRACT

NR4A family orphan nuclear receptors are an important class of transcription factors for development and homeostasis of dopaminergic neurons that also inhibit expression of inflammatory genes in glial cells. The identification of NR4A2 (Nurr1) as a suppressor of nuclear factor κB (NF-κB)-related neuroinflammatory genes in microglia and astrocytes suggests that this receptor could be a target for pharmacologic intervention in neurologic disease, but compounds that promote this activity are lacking. Selected diindolylmethane compounds (C-DIMs) have been shown to activate or inactivate nuclear receptors, including Nurr1, in cancer cells and also suppress astrocyte inflammatory signaling in vitro. Based upon these data, we postulated that C-DIM12 [1,1-bis(3'-indolyl)-1-(p-chlorophenyl) methane] would suppress inflammatory signaling in microglia by a Nurr1-dependent mechanism. C-DIM12 inhibited lipopolysaccharide (LPS)-induced expression of NF-κB-regulated genes in BV-2 microglia including nitric oxide synthase (NOS2), interleukin-6 (IL-6), and chemokine (C-C motif) ligand 2 (CCL2), and the effects were attenuated by Nurr1-RNA interference. Additionally, C-DIM12 decreased NF-κB activation in NF-κB-GFP (green fluorescent protein) reporter cells and enhanced nuclear translocation of Nurr1 primary microglia. Chromatin immunoprecipitation assays indicated that C-DIM12 decreased lipopolysaccharide-induced p65 binding to the NOS2 promoter and concurrently enhanced binding of Nurr1 to the p65-binding site. Consistent with these findings, C-DIM12 also stabilized binding of the Corepressor for Repressor Element 1 Silencing Transcription Factor (CoREST) and the Nuclear Receptor Corepressor 2 (NCOR2). Collectively, these data identify C-DIM12 as a modulator of Nurr1 activity that results in inhibition of NF-κB-dependent gene expression in glial cells by stabilizing nuclear corepressor proteins, which reduces binding of p65 to inflammatory gene promoters.


Subject(s)
Indoles/pharmacology , Microglia/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Cell Line , Co-Repressor Proteins , Cytokines/genetics , Cytokines/metabolism , Gene Expression , Humans , Lipopolysaccharides/pharmacology , Mice , Microglia/immunology , Microglia/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Promoter Regions, Genetic , Protein Transport , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic
10.
J Virol ; 88(3): 1771-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24257615

ABSTRACT

Alphaviruses are mosquito-borne viruses that cause significant disease in animals and humans. Western equine encephalitis virus (WEEV) and eastern equine encephalitis virus (EEEV), two New World alphaviruses, can cause fatal encephalitis, and EEEV is a select agent of concern in biodefense. However, we have no antiviral therapies against alphaviral disease, and current vaccine strategies target only a single alphavirus species. In an effort to develop new tools for a broader response to outbreaks, we designed and tested a novel alphavirus vaccine comprised of cationic lipid nucleic acid complexes (CLNCs) and the ectodomain of WEEV E1 protein (E1ecto). Interestingly, we found that the CLNC component, alone, had therapeutic efficacy, as it increased survival of CD-1 mice following lethal WEEV infection. Immunization with the CLNC-WEEV E1ecto mixture (lipid-antigen-nucleic acid complexes [LANACs]) using a prime-boost regimen provided 100% protection in mice challenged with WEEV subcutaneously, intranasally, or via mosquito. Mice immunized with LANACs mounted a strong humoral immune response but did not produce neutralizing antibodies. Passive transfer of serum from LANAC E1ecto-immunized mice to nonimmune CD-1 mice conferred protection against WEEV challenge, indicating that antibody is sufficient for protection. In addition, the LANAC E1ecto immunization protocol significantly increased survival of mice following intranasal or subcutaneous challenge with EEEV. In summary, our LANAC formulation has therapeutic potential and is an effective vaccine strategy that offers protection against two distinct species of alphavirus irrespective of the route of infection. We discuss plausible mechanisms as well the potential utility of our LANAC formulation as a pan-alphavirus vaccine.


Subject(s)
Antigens, Viral/immunology , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Western Equine/physiology , Encephalomyelitis, Equine/prevention & control , Liposomes/immunology , Nucleic Acids/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Antibodies, Viral/immunology , Antigens, Viral/administration & dosage , Antigens, Viral/chemistry , Antigens, Viral/genetics , Encephalitis Virus, Eastern Equine/immunology , Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/immunology , Encephalomyelitis, Equine/virology , Female , Humans , Immunization , Liposomes/administration & dosage , Liposomes/chemistry , Mice , Nucleic Acids/administration & dosage , Nucleic Acids/chemistry , Viral Proteins/administration & dosage , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/chemistry , Viral Vaccines/genetics
11.
PLoS One ; 8(3): e60427, 2013.
Article in English | MEDLINE | ID: mdl-23544138

ABSTRACT

Western equine encephalitis virus (WEEV) is a naturally occurring recombinant virus derived from ancestral Sindbis and Eastern equine encephalitis viruses. We previously showed that infection by WEEV isolates McMillan (McM) and IMP-181 (IMP) results in high (∼90-100%) and low (0%) mortality, respectively, in outbred CD-1 mice when virus is delivered by either subcutaneous or aerosol routes. However, relatively little is known about specific virulence determinants of WEEV. We previously observed that IMP infected Culex tarsalis mosquitoes at a high rate (app. 80%) following ingestion of an infected bloodmeal but these mosquitoes were infected by McM at a much lower rate (10%). To understand the viral role in these phenotypic differences, we characterized the pathogenic phenotypes of McM/IMP chimeras. Chimeras encoding the E2 of McM on an IMP backbone (or the reciprocal) had the most significant effect on infection phenotypes in mice or mosquitoes. Furthermore, exchanging the arginine, present on IMP E2 glycoprotein at position 214, for the glutamine present at the same position on McM, ablated mouse mortality. Curiously, the reciprocal exchange did not confer mouse virulence to the IMP virus. Mosquito infectivity was also determined and significantly, one of the important loci was the same as the mouse virulence determinant identified above. Replacing either IMP E2 amino acid 181 or 214 with the corresponding McM amino acid lowered mosquito infection rates to McM-like levels. As with the mouse neurovirulence, reciprocal exchange of amino acids did not confer mosquito infectivity. The identification of WEEV E2 amino acid 214 as necessary for both IMP mosquito infectivity and McM mouse virulence indicates that they are mutually exclusive phenotypes and suggests an explanation for the lack of human or equine WEE cases even in the presence of active transmission.


Subject(s)
Culex/virology , Encephalitis Virus, Western Equine/genetics , Encephalitis Virus, Western Equine/pathogenicity , Encephalomyelitis, Western Equine/virology , Nervous System/pathology , Nervous System/virology , Amino Acid Sequence , Amino Acids/metabolism , Animals , Chickens/virology , Encephalitis Virus, Western Equine/growth & development , Encephalomyelitis, Western Equine/genetics , Humans , Mice , Molecular Sequence Data , Point Mutation/genetics , Sequence Alignment , Subcutaneous Tissue/virology , Viremia , Virulence/genetics
12.
PLoS One ; 8(1): e53462, 2013.
Article in English | MEDLINE | ID: mdl-23301074

ABSTRACT

Western equine encephalitis virus (WEEV; Alphavirus) is a mosquito-borne virus that can cause severe encephalitis in humans and equids. Previous studies have shown that intranasal infection of outbred CD-1 mice with the WEEV McMillan (McM) strain result in high mortality within 4 days of infection. Here in vivo and ex vivo bioluminescence (BLM) imaging was applied on mice intranasally infected with a recombinant McM virus expressing firefly luciferase (FLUC) to track viral neuroinvasion by FLUC detection and determine any correlation between BLM and viral titer. Immunological markers of disease (MCP-1 and IP-10) were measured and compared to wild type virus infection. Histopathology was guided by corresponding BLM images, and showed that neuroinvasion occurred primarily through cranial nerves, mainly in the olfactory tract. Olfactory bulb neurons were initially infected with subsequent spread of the infection into different regions of the brain. WEEV distribution was confirmed by immunohistochemistry as having marked neuronal infection but very few infected glial cells. Axons displayed infection patterns consistent with viral dissemination along the neuronal axis. The trigeminal nerve served as an additional route of neuroinvasion showing significant FLUC expression within the brainstem. The recombinant virus WEEV.McM.FLUC had attenuated replication kinetics and induced a weaker immunological response than WEEV.McM but produced comparable pathologies. Immunohistochemistry staining for FLUC and WEEV antigen showed that transgene expression was present in all areas of the CNS where virus was observed. BLM provides a quantifiable measure of alphaviral neural disease progression and a method for evaluating antiviral strategies.


Subject(s)
Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/virology , Luminescent Measurements/methods , Neurons/metabolism , Animals , Antiviral Agents/pharmacology , Brain/pathology , Brain/virology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Equine/genetics , Genes, Reporter , Immunohistochemistry , Luciferases/genetics , Luciferases/metabolism , Mice , Neuroglia/virology , Olfactory Bulb/virology , Time Factors , Transgenes
13.
Antiviral Res ; 87(2): 195-203, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20452378

ABSTRACT

Having recently characterized a CD-1 outbred mouse model of pathogenesis for Western equine encephalitis virus, we examined the possible protective effects of cationic liposome-DNA complexes (CLDCs) against encephalitic arboviral infection. In this investigation, mice were pre-treated, co-treated, or post-treated with CLDC then challenged with a subcutaneous or aerosol dose of the highly virulent WEEV-McMillan strain, lethal in mice 4-5 days after inoculation. Pre-treatment with CLDCs provided a significant protective effect in mice, which was reflected in significantly increased survival rates. Further, in some instances a therapeutic effect of CLDC administration up to 12h after WEEV challenge was observed. Mice treated with CLDC had significantly increased serum IFN-gamma, TNF-alpha, and IL-12, suggesting a strong Th1-biased antiviral activation of the innate immune system. In virus-infected animals, large increases in production of IFN-gamma, TNF-alpha, MCP-1, IL-12, and IL-10 in the brain were observed by 72h after infection, consistent with neuroinvasion and viral replication in the CNS. These results indicate that strong non-specific activation of innate immunity with an immune therapeutic such as CLDC is capable of eliciting significant protective immunity against a rapidly lethal strain of WEEV and suggest a possible prophylactic option for exposed individuals.


Subject(s)
DNA/administration & dosage , Encephalitis Virus, Western Equine/immunology , Encephalomyelitis, Equine/drug therapy , Encephalomyelitis, Equine/prevention & control , Immunologic Factors/administration & dosage , Immunotherapy/methods , Liposomes/administration & dosage , Animals , Blood/immunology , Brain/immunology , Cytokines/analysis , Cytokines/blood , Disease Models, Animal , Drug Carriers/administration & dosage , Encephalitis Virus, Western Equine/genetics , Encephalomyelitis, Equine/immunology , Female , Mice , Survival Analysis
14.
BMC Microbiol ; 9: 49, 2009 Mar 05.
Article in English | MEDLINE | ID: mdl-19265532

ABSTRACT

BACKGROUND: Arthropod-borne viruses (arboviruses) can persistently infect and cause limited damage to mosquito vectors. RNA interference (RNAi) is a mosquito antiviral response important in restricting RNA virus replication and has been shown to be active against some arboviruses. The goal of this study was to use a recombinant Sindbis virus (SINV; family Togaviridae; genus Alphavirus) that expresses B2 protein of Flock House virus (FHV; family Nodaviridae; genus Alphanodavirus), a protein that inhibits RNAi, to determine the effects of linking arbovirus infection with RNAi inhibition. RESULTS: B2 protein expression from SINV (TE/3'2J) inhibited the accumulation of non-specific small RNAs in Aedes aegypti mosquito cell culture and virus-specific small RNAs both in infected cell culture and Ae. aegypti mosquitoes. More viral genomic and subgenomic RNA accumulated in cells and mosquitoes infected with TE/3'2J virus expressing B2 (TE/3'2J/B2) compared to TE/3'2J and TE/3'2J virus expressing GFP. TE/3'2J/B2 exhibited increased infection rates, dissemination rates, and infectious virus titers in mosquitoes following oral bloodmeal. Following infectious oral bloodmeal, significantly more mosquitoes died when TE/3'2J/B2 was ingested. The virus was 100% lethal following intrathoracic inoculation of multiple mosquito species and lethality was dose-dependent in Ae. aegypti. CONCLUSION: We show that RNAi is active in Ae. aegypti cell culture and that B2 protein inhibits RNAi in mosquito cells when expressed by a recombinant SINV. Also, SINV more efficiently replicates in mosquito cells when RNAi is inhibited. Finally, TE/3'2J/B2 kills mosquitoes in a dose-dependent manner independent of infection route and mosquito species.


Subject(s)
Aedes/virology , Alphavirus Infections/virology , Alphavirus/pathogenicity , RNA Interference , Virus Replication/genetics , Alphavirus/physiology , Animals , Chlorocebus aethiops , Cricetinae , Gene Expression Regulation, Viral , RNA, Viral/metabolism , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Sindbis Virus/genetics , Sindbis Virus/pathogenicity , Vero Cells
15.
Virol J ; 3: 74, 2006 Sep 06.
Article in English | MEDLINE | ID: mdl-16956415

ABSTRACT

Simian-Human immunodeficiency virus is a chimeric virus which, in rhesus macaques (Macacca mulatta) closely imitates immunodeficiency virus infection in human (HIV). A relatively new way to study pathogenesis of viral infection is to study alterations in host gene expression induced by the virus. SHIV infection with certain strains does not result in clinical signs. We hypothesized that alterations in gene expression relating to the immune system would be present in SHIV-infected animals despite the lack of clinical signs. Splenic tissue from four adult male Indian-origin Rhesus monkeys serologically positive for non-pathogenic SHIV 89.6 was processed by cDNA microarray analysis. Results were compared with the corresponding outcome using splenic tissues from four unexposed adult male Rhesus monkeys. Subsequent gene analysis confirmed statistically significant variations between control and infected samples. Interestingly, SHIV-infected monkeys exhibited altered expression in genes related to apoptosis, signal transduction, T and B lymphocyte activation and importantly, to immune regulation. Although infected animals appeared asymptomatic, our study demonstrated that SHIV-infected monkeys cannot reliably be used in studies of other infectious agents as their baseline gene expression differs from that of normal Rhesus monkeys. The gene expression differences in SHIV-infected animals relative to uninfected animals offer additional clues to the pathogenesis of altered immune function in response to secondary infection.


Subject(s)
Gene Expression Regulation , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/pathogenicity , Animals , DNA, Complementary/genetics , Genetic Variation , Humans , Macaca mulatta/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Reference Values , Simian Immunodeficiency Virus/isolation & purification , Spleen/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...