Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cell Rep ; 42(8): 112946, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37556325

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus responsible for epidemics of neurological disease across the Americas. Low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3) is a recently reported entry receptor for VEEV. Here, using wild-type and Ldlrad3-deficient mice, we define a critical role for LDLRAD3 in controlling steps in VEEV infection, pathogenesis, and neurotropism. Our analysis shows that LDLRAD3 is required for efficient VEEV infection and pathogenesis prior to and after central nervous system invasion. Ldlrad3-deficient mice survive intranasal and intracranial VEEV inoculation and show reduced infection of neurons in different brain regions. As LDLRAD3 is a determinant of pathogenesis and an entry receptor required for VEEV infection of neurons of the brain, receptor-targeted therapies may hold promise as countermeasures.


Subject(s)
Encephalomyelitis, Venezuelan Equine , Receptors, LDL , Animals , Mice , Brain/pathology , Central Nervous System , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/pathology , Receptors, LDL/physiology
2.
J Virol ; 95(15): e0077721, 2021 07 12.
Article in English | MEDLINE | ID: mdl-34011549

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a reemerging arthropod-borne virus causing encephalitis in humans and domesticated animals. VEEV possesses a positive single-stranded RNA genome capped at its 5' end. The capping process is performed by the nonstructural protein nsP1, which bears methyl and guanylyltransferase activities. The capping reaction starts with the methylation of GTP. The generated m7GTP is complexed to the enzyme to form an m7GMP-nsP1 covalent intermediate. The m7GMP is then transferred onto the 5'-diphosphate end of the viral RNA. Here, we explore the specificities of the acceptor substrate in terms of length, RNA secondary structure, and/or sequence. Any diphosphate nucleosides but GDP can serve as acceptors of the m7GMP to yield m7GpppA, m7GpppC, or m7GpppU. We show that capping is more efficient on small RNA molecules, whereas RNAs longer than 130 nucleotides are barely capped by the enzyme. The structure and sequence of the short, conserved stem-loop, downstream to the cap, is an essential regulatory element for the capping process. IMPORTANCE The emergence, reemergence, and expansion of alphaviruses (genus of the family Togaviridae) are a serious public health and epizootic threat. Venezuelan equine encephalitis virus (VEEV) causes encephalitis in human and domesticated animals, with a mortality rate reaching 80% in horses. To date, no efficient vaccine or safe antivirals are available for human use. VEEV nonstructural protein 1 (nsP1) is the viral capping enzyme characteristic of the Alphavirus genus. nsP1 catalyzes methyltransferase and guanylyltransferase reactions, representing a good therapeutic target. In the present report, we provide insights into the molecular features and specificities of the cap acceptor substrate for the guanylylation reaction.


Subject(s)
Encephalitis Virus, Venezuelan Equine/genetics , RNA Caps/genetics , RNA, Viral/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics , Animals , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/virology , Horses , Humans , Methyltransferases/metabolism , Nucleic Acid Conformation , Nucleotidyltransferases/metabolism , Viral Nonstructural Proteins/genetics
3.
PLoS Negl Trop Dis ; 15(3): e0009306, 2021 03.
Article in English | MEDLINE | ID: mdl-33788849

ABSTRACT

Venezuelan Equine Encephalitis Virus (VEEV) is a major biothreat agent that naturally causes outbreaks in humans and horses particularly in tropical areas of the western hemisphere, for which no antiviral therapy is currently available. The host response to VEEV and the cellular factors this alphavirus hijacks to support its effective replication or evade cellular immune responses are largely uncharacterized. We have previously demonstrated tremendous cell-to-cell heterogeneity in viral RNA (vRNA) and cellular transcript levels during flaviviral infection using a novel virus-inclusive single-cell RNA-Seq approach. Here, we used this unbiased, genome-wide approach to simultaneously profile the host transcriptome and vRNA in thousands of single cells during infection of human astrocytes with the live-attenuated vaccine strain of VEEV (TC-83). Host transcription was profoundly suppressed, yet "superproducer cells" with extremely high vRNA abundance emerged during the first viral life cycle and demonstrated an altered transcriptome relative to both uninfected cells and cells with high vRNA abundance harvested at later time points. Additionally, cells with increased structural-to-nonstructural transcript ratio exhibited upregulation of intracellular membrane trafficking genes at later time points. Loss- and gain-of-function experiments confirmed pro- and antiviral activities in both vaccine and virulent VEEV infections among the products of transcripts that positively or negatively correlated with vRNA abundance, respectively. Lastly, comparison with single cell transcriptomic data from other viruses highlighted common and unique pathways perturbed by infection across evolutionary scales. This study provides a high-resolution characterization of the VEEV (TC-83)-host interplay, identifies candidate targets for antivirals, and establishes a comparative single-cell approach to study the evolution of virus-host interactions.


Subject(s)
Biological Transport/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/pathology , Host-Pathogen Interactions/genetics , Transcription, Genetic/genetics , Virus Internalization , Animals , Antibodies, Viral/immunology , Astrocytes/virology , Cell Line , Chlorocebus aethiops , Cricetinae , Encephalitis Virus, Venezuelan Equine/immunology , Gene Expression Regulation, Viral/genetics , Horses , Humans , RNA, Viral/genetics , Single-Cell Analysis , Vaccines, Attenuated/immunology , Vero Cells , Virus Replication/physiology
4.
Antiviral Res ; 182: 104875, 2020 10.
Article in English | MEDLINE | ID: mdl-32755661

ABSTRACT

Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV) are mosquito-borne viruses in the Americas that cause central nervous system (CNS) disease in humans and equids. In this study, we directly characterized the pathogenesis of VEEV, EEEV, and WEEV in cynomolgus macaques following subcutaneous exposure because this route more closely mimics natural infection via mosquito transmission or by an accidental needle stick. Our results highlight how EEEV is significantly more pathogenic compared to VEEV similarly to what is observed in humans. Interestingly, EEEV appears to be just as neuropathogenic by subcutaneous exposure as it was in previously completed aerosol exposure studies. In contrast, subcutaneous exposure of cynomolgus macaques with WEEV caused limited disease and is contradictory to what has been reported for aerosol exposure. Several differences in viremia, hematology, or tissue tropism were noted when animals were exposed subcutaneously compared to prior aerosol exposure studies. This study provides a more complete picture of the pathogenesis of the encephalitic alphaviruses and highlights how further defining the neuropathology of these viruses could have important implications for the development of medical countermeasures for the neurovirulent alphaviruses.


Subject(s)
Encephalitis Virus, Eastern Equine/pathogenicity , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalitis Virus, Western Equine/pathogenicity , Encephalomyelitis, Equine/pathology , Encephalomyelitis, Venezuelan Equine/pathology , Macaca fascicularis/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Male , Virus Replication
5.
Virology ; 539: 121-128, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31733451

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a neurotropic virus that causes significant disease in both humans and equines. Here we characterized the impact of VEEV on signaling pathways regulating cell death in human primary astrocytes. VEEV productively infected primary astrocytes and caused an upregulation of early growth response 1 (EGR1) gene expression at 9 and 18 h post infection. EGR1 induction was dependent on extracellular signal-regulated kinase1/2 (ERK1/2) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), but not on p38 mitogen activated protein kinase (MAPK) or phosphoinositide 3-kinase (PI3K) signaling. Knockdown of EGR1 significantly reduced VEEV-induced apoptosis and impacted viral replication. Knockdown of ERK1/2 or PERK significantly reduced EGR1 gene expression, dramatically reduced viral replication, and increased cell survival as well as rescued cells from VEEV-induced apoptosis. These data indicate that EGR1 activation and subsequent cell death are regulated through ERK and PERK pathways in VEEV infected primary astrocytes.


Subject(s)
Cell Death , Early Growth Response Protein 1/genetics , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/virology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , eIF-2 Kinase/metabolism , Apoptosis , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Cell Survival , Cells, Cultured , Early Growth Response Protein 1/metabolism , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/pathology , Gene Expression , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Signal Transduction , Virus Replication , eIF-2 Kinase/genetics
6.
PLoS One ; 9(2): e86745, 2014.
Article in English | MEDLINE | ID: mdl-24586253

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) belongs to the genus Alphavirus, family Togaviridae. VEEV infection is characterized by extensive inflammation and studies from other laboratories implicated an involvement of the NF-κB cascade in the in vivo pathology. Initial studies indicated that at early time points of VEEV infection, the NF-κB complex was activated in cells infected with the TC-83 strain of VEEV. One upstream kinase that contributes to the phosphorylation of p65 is the IKKß component of the IKK complex. Our previous studies with Rift valley fever virus, which exhibited early activation of the NF-κB cascade in infected cells, had indicated that the IKKß component underwent macromolecular reorganization to form a novel low molecular weight form unique to infected cells. This prompted us to investigate if the IKK complex undergoes a comparable macromolecular reorganization in VEEV infection. Size-fractionated VEEV infected cell extracts indicated a macromolecular reorganization of IKKß in VEEV infected cells that resulted in formation of lower molecular weight complexes. Well-documented inhibitors of IKKß function, BAY-11-7082, BAY-11-7085 and IKK2 compound IV, were employed to determine whether IKKß function was required for the production of infectious progeny virus. A decrease in infectious viral particles and viral RNA copies was observed with inhibitor treatment in the attenuated and virulent strains of VEEV infection. In order to further validate the requirement of IKKß for VEEV replication, we over-expressed IKKß in cells and observed an increase in viral titers. In contrast, studies carried out using IKKß(-/-) cells demonstrated a decrease in VEEV replication. In vivo studies demonstrated that inhibitor treatment of TC-83 infected mice increased their survival. Finally, proteomics studies have revealed that IKKß may interact with the viral protein nsP3. In conclusion, our studies have revealed that the host IKKß protein may be critically involved in VEEV replication.


Subject(s)
Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/metabolism , I-kappa B Kinase/metabolism , Animals , Cell Line , Down-Regulation/drug effects , Encephalomyelitis, Venezuelan Equine/pathology , Guinea Pigs , Host-Pathogen Interactions , Humans , Mice , NF-kappa B/metabolism , Neurons/virology , Nitriles/pharmacology , Signal Transduction/drug effects , Sulfones/pharmacology , Viral Load/drug effects , Viral Proteins/metabolism , Virus Replication/drug effects
7.
Vet Microbiol ; 167(1-2): 145-50, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-23968890

ABSTRACT

Equine encephalids have high mortality rates and represent a significant zoonotic public health threat. Of these the most pathogenic viruses to equids are the alphaviruses in the family Togaviridae. The focus of this review Venezualen equine encephalitis virus (VEEV) has caused the most widespread and recent epidemic outbreaks of disease. Circulation in naturally occuring rodent-mosquito cycles, results in viral spread to both human and equine populations. However, equines develop a high titer viremia and can transmit the virus back to mosquito populations. As such, the early recognition and control of viral infection in equine populations is strongly associated with prevention of epidemic spread of the virus and limiting of disease incidence in human populations. This review will address identification and pathogenesis of VEEV in equids vaccination and treatment options, and current research for drug and vaccine development.


Subject(s)
Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/virology , Horse Diseases/virology , Animals , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/prevention & control , Encephalomyelitis, Venezuelan Equine/transmission , Horse Diseases/prevention & control , Horse Diseases/transmission , Horses , Humans , Zoonoses/virology
8.
Vaccine ; 30(27): 4095-105, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22525795

ABSTRACT

TC83 is a human vaccine with investigational new drug status and is used as a prototype Venezuelan equine encephalitis virus for pathogenesis and antiviral research. Differing from other experimental models, the virus causes high titer infection in the brain and 90-100% mortality in the C3H/HeN murine model. To better characterize the susceptibility to disease development in C3H/HeN mice, we have analyzed the gene transcriptomes and cytokine production in the brains of infected mice. Our analysis indicated the potential importance of natural killer cells in the encephalitic disease development. This paper describes for the first time a pathogenic role for natural killer cells in VEEV encephalitis.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/pathology , Killer Cells, Natural/immunology , Animals , Brain/pathology , Brain/virology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Venezuelan Equine/mortality , Gene Expression Profiling , Mice , Mice, Inbred C3H , Survival Analysis
9.
Vaccine ; 29(50): 9345-51, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-22001884

ABSTRACT

Rapid and accurate identification of disease progression are key factors in testing novel vaccines and antivirals against encephalitic alphaviruses. Typical efficacy studies utilize a large number of animals and severe morbidity or mortality as an endpoint. New technologies provide a means to reduce and refine the animal use as proposed in Hume's 3Rs (replacement, reduction, refinement) described by Russel and Burch. In vivo imaging systems (IVIS) and bioluminescent enzyme technologies accomplish the reduction of animal requirements while shortening the experimental time and improving the accuracy in localizing active virus replication. In the case of murine models of viral encephalitis in which central nervous system (CNS) viral invasion occurs rapidly but the disease development is relatively slow, we visualized the initial brain infection and enhance the data collection process required for efficacy studies on antivirals or vaccines that are aimed at preventing brain infection. Accordingly, we infected mice through intranasal inoculation with the genetically modified pathogen, Venezuelan equine encephalitis, which expresses a luciferase gene. In this study, we were able to identify the invasion of the CNS at least 3 days before any clinical signs of disease, allowing for reduction of animal morbidity providing a humane means of disease and vaccine research while obtaining scientific data accurately and more rapidly. Based on our data from the imaging model, we confirmed the usefulness of this technology in preclinical research by demonstrating the efficacy of Ampligen, a TLR-3 agonist, in preventing CNS invasion.


Subject(s)
Brain/virology , Central Nervous System Diseases/virology , Encephalomyelitis, Venezuelan Equine/pathology , Imaging, Three-Dimensional/methods , Neuroimaging/methods , Animals , Antiviral Agents/pharmacology , Central Nervous System Diseases/prevention & control , Disease Models, Animal , Encephalomyelitis, Venezuelan Equine/drug therapy , Female , Mice , Mice, Inbred ICR , Poly I-C/pharmacology , Poly U/pharmacology
10.
J Virol ; 85(20): 10682-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21849461

ABSTRACT

Venezuelan equine encephalitis (VEE) virus is a mosquito-borne alphavirus associated with sporadic outbreaks in human and equid populations in the Western Hemisphere. After the bite of an infected mosquito, the virus initiates a biphasic disease: a peripheral phase with viral replication in lymphoid and myeloid tissues, followed by a neurotropic phase with infection of central nervous system (CNS) neurons, causing neuropathology and in some cases fatal encephalitis. The mechanisms allowing VEE virus to enter the CNS are currently poorly understood. Previous data have shown that the virus gains access to the CNS by infecting olfactory sensory neurons in the nasal mucosa of mice. However, at day 5 after inoculation, the infection of the brain is multifocal, indicating that virus particles are able to cross the blood-brain barrier (BBB). To better understand the role of the BBB during VEE virus infection, we used a well-characterized mouse model system. Using VEE virus replicon particles (VRP), we modeled the early events of neuroinvasion, showing that the replication of VRP in the nasal mucosa induced the opening of the BBB, allowing peripherally administered VRP to invade the brain. Peripheral VEE virus infection was characterized by a biphasic opening of the BBB. Further, inhibition of BBB opening resulted in a delayed viral neuroinvasion and pathogenesis. Overall, these results suggest that VEE virus initially enters the CNS through the olfactory pathways and initiates viral replication in the brain, which induces the opening of the BBB, allowing a second wave of invading virus from the periphery to enter the brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Blood-Brain Barrier/virology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/virology , Animals , Disease Models, Animal , Mice , Mice, Inbred BALB C , Olfactory Receptor Neurons/virology , Rodent Diseases/pathology , Rodent Diseases/virology
11.
Virol J ; 8: 197, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21529366

ABSTRACT

BACKGROUND: Neuroinvasion of Venezuelan equine encephalitis virus (VEEV) and subsequent initiation of inflammation in the brain plays a crucial role in the outcome of VEEV infection in mice. Adhesion molecules expressed on microvascular endothelial cells in the brain have been implicated in the modulation of the blood brain barrier (BBB) and inflammation in brain but their role in VEEV pathogenesis is not very well understood. In this study, we evaluated the expression of extracellular matrix and adhesion molecules genes in the brain of VEEV infected mice. FINDINGS: Several cell to cell adhesion molecules and extracellular matrix protein genes such as ICAM-1, VCAM-1, CD44, Cadherins, integrins, MMPs and Timp1 were differentially regulated post-VEEV infection. ICAM-1 knock-out (IKO) mice infected with VEEV had markedly reduced inflammation in the brain and demonstrated a delay in the onset of clinical symptoms of disease. A differential regulation of inflammatory genes was observed in the IKO mice brain compared to their WT counterparts. CONCLUSIONS: These results improve our present understanding of VEEV induced inflammation in mouse brain.


Subject(s)
Brain/pathology , Brain/virology , Cell Adhesion Molecules/biosynthesis , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/pathology , Inflammation/pathology , Animals , Disease Models, Animal , Encephalomyelitis, Venezuelan Equine/virology , Gene Expression Profiling , Histocytochemistry , Immunohistochemistry , Mice , Mice, Knockout , Microscopy , Rodent Diseases/pathology , Rodent Diseases/virology
12.
Clin Vaccine Immunol ; 18(5): 707-16, 2011 May.
Article in English | MEDLINE | ID: mdl-21450977

ABSTRACT

We evaluated the immunogenicity and protective efficacy of a DNA vaccine expressing codon-optimized envelope glycoprotein genes of Venezuelan equine encephalitis virus (VEEV) when delivered by intramuscular electroporation. Mice vaccinated with the DNA vaccine developed robust VEEV-neutralizing antibody responses that were comparable to those observed after administration of the live-attenuated VEEV vaccine TC-83 and were completely protected from a lethal aerosol VEEV challenge. The DNA vaccine also elicited strong neutralizing antibody responses in rabbits that persisted at high levels for at least 6 months and could be boosted by a single additional electroporation administration of the DNA performed approximately 6 months after the initial vaccinations. Cynomolgus macaques that received the vaccine by intramuscular electroporation developed substantial neutralizing antibody responses and after an aerosol challenge had no detectable serum viremia and had reduced febrile reactions, lymphopenia, and clinical signs of disease compared to those of negative-control macaques. Taken together, our results demonstrate that this DNA vaccine provides a potent means of protecting against VEEV infections and represents an attractive candidate for further development.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Vaccines, DNA/immunology , Viral Vaccines/immunology , Animals , Disease Models, Animal , Electroporation , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/pathology , Female , Fever/prevention & control , Glycoproteins/genetics , Glycoproteins/immunology , Lymphopenia/prevention & control , Macaca , Male , Mice , Mice, Inbred BALB C , Rabbits , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Time Factors , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viremia/prevention & control
13.
J Virol ; 84(9): 4556-68, 2010 May.
Article in English | MEDLINE | ID: mdl-20181704

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a mosquito-borne RNA virus of the genus Alphavirus that is responsible for a significant disease burden in Central and South America through sporadic outbreaks into human and equid populations. For humans, 2 to 4% of cases are associated with encephalitis, and there is an overall case mortality rate of approximately 1%. In mice, replication of the virus within neurons of the central nervous system (CNS) leads to paralyzing, invariably lethal encephalomyelitis. However, mice infected with certain attenuated mutants of the virus are able to control the infection within the CNS and recover. To better define what role T cell responses might be playing in this process, we infected B cell-deficient microMT mice with a VEEV mutant that induces mild, sublethal illness in immune competent mice. Infected microMT mice rapidly developed the clinical signs of severe paralyzing encephalomyelitis but were eventually able to control the infection and recover fully from clinical illness. Recovery in this system was T cell dependent and associated with a dramatic reduction in viral titers within the CNS, followed by viral persistence in the brain. Further comparison of the relative roles of T cell subpopulations within this system revealed that CD4(+) T cells were better producers of gamma interferon (IFN-gamma) than CD8(+) T cells and were more effective at controlling VEEV within the CNS. Overall, these results suggest that T cells, especially CD4(+) T cells, can successfully control VEEV infection within the CNS and facilitate recovery from a severe viral encephalomyelitis.


Subject(s)
Antibodies, Viral/immunology , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/immunology , T-Lymphocytes/immunology , Animals , Brain/virology , Encephalomyelitis, Venezuelan Equine/pathology , Female , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology , Viral Load
14.
J Virol ; 83(9): 4275-86, 2009 May.
Article in English | MEDLINE | ID: mdl-19225006

ABSTRACT

Venezuelan equine encephalitis virus (VEE) replicon particles (VRP) were used to model the initial phase of VEE-induced encephalitis in the mouse brain. VRP can target and infect cells as VEE, but VRP do not propagate beyond the first infected cell due to the absence of the structural genes. Direct intracranial inoculation of VRP into mice induced acute encephalitis with signs similar to the neuronal phase of wild-type VEE infection and other models of virus-induced encephalitis. Using the previously established VRP-mRNP tagging system, a new method to distinguish the host responses in infected cells from those in uninfected bystander cell populations, we detected a robust and rapid innate immune response in the central nervous system (CNS) by infected neurons and uninfected bystander cells. Moreover, this innate immune response in the CNS compromised blood-brain barrier integrity, created an inflammatory response, and directed an adaptive immune response characterized by proliferation and activation of microglia cells and infiltration of inflammatory monocytes, in addition to CD4(+) and CD8(+) T lymphocytes. Taken together, these data suggest that a naïve CNS has an intrinsic potential to induce an innate immune response that could be crucial to the outcome of the infection by determining the composition and dynamics of the adaptive immune response. Furthermore, these results establish a model for neurotropic virus infection to identify host and viral factors that contribute to invasion of the brain, the mechanism(s) whereby the adaptive immune response can clear the infection, and the role of the host innate response in these processes.


Subject(s)
Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/growth & development , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/virology , Virion/growth & development , Animals , Cell Proliferation , Cytokines/metabolism , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/pathology , Female , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred BALB C , Microglia/metabolism , Microglia/pathology , RNA, Viral/genetics , Virion/genetics
16.
BMC Genomics ; 9: 289, 2008 Jun 16.
Article in English | MEDLINE | ID: mdl-18558011

ABSTRACT

BACKGROUND: Neurovirulent Venezuelan equine encephalitis virus (VEEV) causes lethal encephalitis in equines and is transmitted to humans by mosquitoes. VEEV is highly infectious when transmitted by aerosol and has been developed as a bio-warfare agent, making it an important pathogen to study from a military and civilian standpoint. Molecular mechanisms of VEE pathogenesis are poorly understood. To study these, the gene expression profile of VEEV infected mouse brains was investigated. Changes in gene expression were correlated with histological changes in the brain. In addition, a molecular framework of changes in gene expression associated with progression of the disease was studied. RESULTS: Our results demonstrate that genes related to important immune pathways such as antigen presentation, inflammation, apoptosis and response to virus (Cxcl10, CxCl11, Ccl5, Ifr7, Ifi27 Oas1b, Fcerg1,Mif, Clusterin and MHC class II) were upregulated as a result of virus infection. The number of over-expressed genes (>1.5-fold level) increased as the disease progressed (from 197, 296, 400, to 1086 at 24, 48, 72 and 96 hours post infection, respectively). CONCLUSION: Identification of differentially expressed genes in brain will help in the understanding of VEEV-induced pathogenesis and selection of biomarkers for diagnosis and targeted therapy of VEEV-induced neurodegeneration.


Subject(s)
Brain/immunology , Brain/pathology , Encephalomyelitis, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/immunology , Immunity/genetics , Inflammation/genetics , Animals , Antigen Presentation/genetics , Antigens/immunology , Apoptosis/genetics , Brain/metabolism , Encephalomyelitis, Venezuelan Equine/pathology , Eosine Yellowish-(YS)/metabolism , Gene Expression Profiling , Hematoxylin/metabolism , Immunohistochemistry , Male , Mice , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate
17.
Am J Trop Med Hyg ; 78(1): 83-92, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18187789

ABSTRACT

A vectored vaccine based on equine herpesvirus type 1 (EHV-1) was generated as an alternative for safe and efficient prophylaxis against Venezuelan equine encephalitis virus (VEEV) infection. Two-step (en passant) Red mutagenesis was used to insert VEEV structural genes into an infectious clone of EHV-1 vaccine strain RacH. The recombinant virus, rH_VEEV, efficiently and stably expressed VEEV structural proteins as detected by various antibodies, including a conformation-dependent monoclonal antibody to envelope glycoprotein E2. In addition, rH_VEEV was indistinguishable from parental bacterial artificial chromosome-derived virus with respect to growth properties in cultured cells. Immunization of mice with the vectored vaccine conferred full protection against lethal challenge infection using VEEV strain ZPC738 in the absence of neutralizing antibodies and in a dose-dependent manner. Analyses of IgG responses demonstrated production of VEEV-specific IgG1 and total IgG antibodies after vaccination, indicating that protection was dependent on either cytotoxic T cell responses or antibody-mediated protection unrelated to neutralizing activity.


Subject(s)
Antibodies, Viral , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Vaccines, Synthetic/administration & dosage , Viral Vaccines/administration & dosage , Animals , Brain/virology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/pathology , Female , Horses , Mice , Recombination, Genetic , Vaccination/veterinary , Viral Structural Proteins/genetics , Virus Replication
18.
J Virol ; 81(24): 13552-65, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17913819

ABSTRACT

The encephalitogenic New World alphaviruses, including Venezuelan (VEEV), eastern (EEEV), and western equine encephalitis viruses, constitute a continuing public health threat in the United States. They circulate in Central, South, and North America and have the ability to cause fatal disease in humans and in horses and other domestic animals. We recently demonstrated that these viruses have developed the ability to interfere with cellular transcription and use it as a means of downregulating a cellular antiviral response. The results of the present study suggest that the N-terminal, approximately 35-amino-acid-long peptide of VEEV and EEEV capsid proteins plays the most critical role in the downregulation of cellular transcription and development of a cytopathic effect. The identified VEEV-specific peptide C(VEE)33-68 includes two domains with distinct functions: the alpha-helix domain, helix I, which is critically involved in supporting the balance between the presence of the protein in the cytoplasm and nucleus, and the downstream peptide, which might contain a functional nuclear localization signal(s). The integrity of both domains not only determines the intracellular distribution of the VEEV capsid but is also essential for direct capsid protein functioning in the inhibition of transcription. Our results suggest that the VEEV capsid protein interacts with the nuclear pore complex, and this interaction correlates with the protein's ability to cause transcriptional shutoff and, ultimately, cell death. The replacement of the N-terminal fragment of the VEEV capsid by its Sindbis virus-specific counterpart in the VEEV TC-83 genome does not affect virus replication in vitro but reduces cytopathogenicity and results in attenuation in vivo. These findings can be used in designing a new generation of live, attenuated, recombinant vaccines against the New World alphaviruses.


Subject(s)
Capsid Proteins/metabolism , Capsid Proteins/pharmacology , Encephalitis Virus, Venezuelan Equine/pathogenicity , Proteins/metabolism , Transcription, Genetic/drug effects , Animals , Capsid Proteins/genetics , Cell Survival , Cricetinae , Cytopathogenic Effect, Viral , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/mortality , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/virology , Female , Immunization , Mice , Mutation , Proteins/genetics
19.
Virology ; 367(2): 307-23, 2007 Oct 25.
Article in English | MEDLINE | ID: mdl-17610927

ABSTRACT

We evaluated the safety and immunogenicity of a chimeric alphavirus vaccine candidate in mice with selective immunodeficiencies. This vaccine candidate was highly attenuated in mice with deficiencies in the B and T cell compartments, as well as in mice with deficient gamma-interferon responsiveness. However, the level of protection varied among the strains tested. Wild type mice were protected against lethal VEEV challenge. In contrast, alpha/beta (alphabeta) TCR-deficient mice developed lethal encephalitis following VEEV challenge, while mice deficient in gamma/delta (gammadelta) T cells were protected. Surprisingly, the vaccine potency was diminished by 50% in animals lacking interferon-gamma receptor alpha chain (R1)-chain and a minority of vaccinated immunoglobulin heavy chain-deficient (microMT) mice survived challenge, which suggests that neutralizing antibody may not be absolutely required for protection. Prolonged replication of encephalitic VEEV in the brain of pre-immunized mice is not lethal and adoptive transfer experiments indicate that CD3(+) T cells are required for protection.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , Viral Vaccines/immunology , Animals , Disease Models, Animal , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Safety , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
20.
Vaccine ; 25(10): 1868-76, 2007 Feb 26.
Article in English | MEDLINE | ID: mdl-17240002

ABSTRACT

A new vaccine, V3526, is a live-attenuated virus derived by site-directed mutagenesis from a virulent clone of the Venezuelan equine encephalitis virus (VEEV) IA/B Trinidad donkey (TrD) strain, intended for human use in protection against Venezuelan equine encephalitis (VEE). Two studies were conducted in horses to evaluate the safety, immunogenicity, ability to boost and protective efficacy of V3526 against challenges of TrD and VEEV IE 64A99. Horses were vaccinated subcutaneously (SC) with 10(7), 10(5), 10(3) or 10(2) plaque-forming units (pfu) of V3526. Control horses were sham immunized. In the first study, challenge viruses (TrD or 64A99) were administered SC 28 days post-vaccination (PV). No viremia and only mild fluctuation in white blood cell counts were observed PV. None of the V3526 vaccinated horses showed clinical signs of disease or pathology of VEE post-challenge (PC). In contrast, control horses challenged SC with 10(4)pfu TrD became viremic and showed classical signs of VEE beginning on Day 3 PC, including elevated body temperature, anorexia, leukopenia and malaise. Moderate to severe encephalitis was found in three of five control horses challenged with TrD. Control horses challenged with 64A99 failed to develop detectable viremia, but did exhibit a brief febrile episode at 1-3 days PC. None of the 10 immunized horses challenged with 64A99 became pyrexic. Twenty four of 25 horses immunized with V3526 in the first study developed serum neutralizing antibody to TrD and 64A99 within 14 days PV. Vaccinations with V3526, at doses as low as 10(2)pfu, were safe and efficacious in protecting horses against a virulent TrD virus challenge. The second study supported that repeat dosing resulted in an increase in serum neutralizing antibody to TrD.


Subject(s)
Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/prevention & control , Horse Diseases/prevention & control , Viral Vaccines/adverse effects , Viral Vaccines/immunology , Animals , Encephalitis Virus, Venezuelan Equine/isolation & purification , Encephalomyelitis, Venezuelan Equine/pathology , Encephalomyelitis, Venezuelan Equine/physiopathology , Female , Histocytochemistry , Horses , Injections, Subcutaneous , Kidney/pathology , Leukocyte Count , Liver/pathology , Lung/pathology , Lymph Nodes/pathology , Male , Myocardium/pathology , Pancreas/pathology , Spleen/pathology , Telencephalon/pathology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology , Viral Plaque Assay , Viremia/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...