Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters











Publication year range
1.
Vet Pathol ; 59(4): 696-706, 2022 07.
Article in English | MEDLINE | ID: mdl-34963403

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019, which ranges from fatal disease in some to mild or subclinical in most affected individuals. Many recovered human patients report persistent respiratory signs; however, lung disease in post-acute infection is poorly understood. Our objective was to describe histologic lung lesions and viral loads following experimental SARS-CoV-2 infection in 11 cats. Microscopic evaluation at 3, 6, 10, or 28 days postinoculation (DPI) identified mild to moderate patchy interstitial pneumonia, bronchiolar epithelial damage, and occlusive histiocytic bronchiolitis. Based on immunohistochemistry, alveolar septal thickening was due to CD204-positive macrophages, fewer B and T lymphocytes, type II pneumocytes, and capillary proliferation with a relative dearth of fibrosis. In blood vessel endothelium, there was reactive hypertrophy or vacuolar degeneration and increased MHC II expression at all time points. Unexpectedly, one cat from the 28 DPI group had severe subacute regionally extensive lymphohistiocytic pneumonia with multifocal consolidation, vasculitis, and alveolar fibrin. Reverse transcriptase-quantitative polymerase chain reaction identified SARS-CoV-2 RNA within the lung at 3 and 6 DPI, and viral RNA was below the limit of detection at 10 and 28 DPI, suggesting that pulmonary lesions persist beyond detection of viral RNA. These findings clarify our comparative understanding of disease induced by SARS-CoV-2 and suggest that cats can serve as an informative model to study post-acute pulmonary sequelae.


Subject(s)
COVID-19 , Cat Diseases , Animals , COVID-19/veterinary , Cat Diseases/pathology , Cats , Humans , Immunohistochemistry , Lung/pathology , RNA, Viral , SARS-CoV-2
2.
PLoS Negl Trop Dis ; 12(11): e0006880, 2018 11.
Article in English | MEDLINE | ID: mdl-30418969

ABSTRACT

The recent emergence of Zika virus (ZIKV) in the Americas coincident with increased caseloads of microcephalic infants and Guillain-Barre syndrome has prompted a flurry of research on ZIKV. Much of the research is difficult to compare or repeat because individual laboratories use different virus isolates, growth conditions, and quantitative assays. Here we obtained three readily available contemporary ZIKV isolates and the prototype Ugandan isolate. We generated stocks of each on Vero mammalian cells (ZIKVmam) and C6/36 mosquito cells (ZIKVmos), determined titers by different assays side-by-side, compared growth characteristics using one-step and multi-step growth curves on Vero and C6/36 cells, and examined plaque phenotype. ZIKV titers consistently peaked earlier on Vero cells than on C6/36 cells. Contemporary ZIKV isolates reached peak titer most quickly in a multi-step growth curve when the amplifying cell line was the same as the titering cell line (e.g., ZIKVmam titered on Vero cells). Growth of ZIKVmam on mosquito cells was particularly delayed. These data suggest that the ability to infect and/or replicate in insect cells is limited after growth in mammalian cells. In addition, ZIKVmos typically had smaller, more homogenous plaques than ZIKVmam in a standard plaque assay. We hypothesized that the plaque size difference represented early adaptation to growth in mammalian cells. We plaque purified representative-sized plaques from ZIKVmos and ZIKVmam. ZIKVmos isolates maintained the initial phenotype while plaques from ZIKVmam isolates became larger with passaging. Our results underscore the importance of the cells used to produce viral stocks and the potential for adaptation with minimal cell passages. In addition, these studies provide a foundation to compare current and emerging ZIKV isolates in vitro and in vivo.


Subject(s)
Adaptation, Physiological , Aedes/cytology , Zika Virus/growth & development , Zika Virus/physiology , Aedes/virology , Animals , Cell Line , Chlorocebus aethiops , Humans , In Vitro Techniques , Phenotype , Vero Cells , Virus Replication , Zika Virus/classification , Zika Virus/isolation & purification
3.
Virology ; 518: 385-397, 2018 05.
Article in English | MEDLINE | ID: mdl-29605685

ABSTRACT

Feline herpes virus type 1 (FHV-1) is widely considered to be the leading cause of ocular disease in cats and has been implicated in upper respiratory tract infections. Little, however is known about interstrain phylogenetic relationships, and details of the genomic structure. For the present study, twenty-six FHV-1 isolates from different cats in animal shelters were collected from eight separate locations in the USA, and the genomes sequenced. Genomic characterization of these isolates includied short sequence repeat (SSR) detection, with fewer SSRs detected, compared to herpes simplex viruses type 1 and 2. For phylogenetic and recombination analysis, 27 previously sequenced isolates of FHV-1 were combined with the 26 strains sequenced for the present study. The overall genomic interstrain genetic distance between all available isolates was 0.093%. Phylogenetic analysis identified four main FHV-1 clades primarily corresponding to geographical collection site. Recombination analysis suggested that interclade recombination has occurred.


Subject(s)
Genetic Variation , Genome, Viral , Phylogeny , Recombination, Genetic , Varicellovirus/classification , Varicellovirus/genetics , Animals , Cat Diseases/virology , Cats , Global Health , Herpesviridae Infections/veterinary , Sequence Analysis, DNA , Varicellovirus/isolation & purification
4.
Insect Biochem Mol Biol ; 95: 1-9, 2018 04.
Article in English | MEDLINE | ID: mdl-29526772

ABSTRACT

Previously, a Sanger-based sialotranscriptome analysis of adult female Culex tarsalis was published based on ∼2000 ESTs. During the elapsed 7.5 years, pyrosequencing has been discontinued and Illumina sequences have increased considerable in size and decreased in price. We here report an Illumina-based sialotranscriptome that allowed finding the missing apyrase from the salivary transcriptome of C. tarsalis, to determine several full-length members of the 34-62 kDa family, when a single EST has been found previously, in addition to identifying many salivary families with lower expression levels that were not detected previously. The use of multiple libraries including salivary glands and carcasses from male and female organisms allowed for an unprecedented insight into the tissue specificity of transcripts, and in this particular case permitting identification of transcripts putatively associated with blood feeding, when exclusive of female salivary glands, or associated with sugar feeding, when transcripts are found upregulated in both male and female glands.


Subject(s)
Culex/metabolism , Insect Proteins/biosynthesis , Sex Characteristics , Sialoglycoproteins/biosynthesis , Animals , Female , Gene Expression Profiling , Male
5.
Genome Announc ; 6(4)2018 Jan 25.
Article in English | MEDLINE | ID: mdl-29371358

ABSTRACT

We report 26 complete genomes of Zika virus (ZIKV) isolated after passaging the Zika virus strain FLR in mosquito (C6/36) and mammalian (Vero) cell lines. The consensus ZIKV genomes we recovered show greater than 99% nucleotide identify with each other and with the FLR strain used as input.

6.
Elife ; 62017 07 31.
Article in English | MEDLINE | ID: mdl-28758638

ABSTRACT

Influenza virus expresses transcripts early in infection and transitions towards genome replication at later time points. This process requires de novo assembly of the viral replication machinery, large ribonucleoprotein complexes (RNPs) composed of the viral polymerase, genomic RNA and oligomeric nucleoprotein (NP). Despite the central role of RNPs during infection, the factors dictating where and when they assemble are poorly understood. Here we demonstrate that human protein kinase C (PKC) family members regulate RNP assembly. Activated PKCδ interacts with the polymerase subunit PB2 and phospho-regulates NP oligomerization and RNP assembly during infection. Consistent with its role in regulating RNP assembly, knockout of PKCδ impairs virus infection by selectively disrupting genome replication. However, primary transcription from pre-formed RNPs deposited by infecting particles is unaffected. Thus, influenza virus exploits host PKCs to regulate RNP assembly, a step required for the transition from primary transcription to genome replication during the infectious cycle.


Subject(s)
Host-Pathogen Interactions , Influenza A Virus, H1N1 Subtype/genetics , Protein Kinase C-delta/genetics , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , Ribonucleoproteins/genetics , Viral Proteins/genetics , Virus Replication , A549 Cells , Animals , Dogs , Gene Expression Regulation , HEK293 Cells , Humans , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Madin Darby Canine Kidney Cells , Models, Molecular , Mutation , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinase C-delta/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/metabolism , Ribonucleoproteins/metabolism , Signal Transduction , Transcription, Genetic , Viral Proteins/metabolism
7.
PLoS Negl Trop Dis ; 11(2): e0005394, 2017 02.
Article in English | MEDLINE | ID: mdl-28187142

ABSTRACT

Half of the human population is at risk of infection by an arthropod-borne virus. Many of these arboviruses, such as West Nile, dengue, and Zika viruses, infect humans by way of a bite from an infected mosquito. This infectious inoculum is insect cell-derived giving the virus particles distinct qualities not present in secondary infectious virus particles produced by infected vertebrate host cells. The insect cell-derived particles differ in the glycosylation of virus structural proteins and the lipid content of the envelope, as well as their induction of cytokines. Thus, in order to accurately mimic the inoculum delivered by arthropods, arboviruses should be derived from arthropod cells. Previous studies have packaged replicon genome in mammalian cells to produce replicon particles, which undergo only one round of infection, but no studies exist packaging replicon particles in mosquito cells. Here we optimized the packaging of West Nile virus replicon genome in mosquito cells and produced replicon particles at high concentration, allowing us to mimic mosquito cell-derived viral inoculum. These particles were mature with similar genome equivalents-to-infectious units as full-length West Nile virus. We then compared the mosquito cell-derived particles to mammalian cell-derived particles in mice. Both replicon particles infected skin at the inoculation site and the draining lymph node by 3 hours post-inoculation. The mammalian cell-derived replicon particles spread from the site of inoculation to the spleen and contralateral lymph nodes significantly more than the particles derived from mosquito cells. This in vivo difference in spread of West Nile replicons in the inoculum demonstrates the importance of using arthropod cell-derived particles to model early events in arboviral infection and highlights the value of these novel arthropod cell-derived replicon particles for studying the earliest virus-host interactions for arboviruses.


Subject(s)
Aedes/virology , Arbovirus Infections/virology , Arboviruses/physiology , Insect Vectors/virology , Replicon , West Nile Fever/virology , West Nile virus/genetics , Animals , Arboviruses/genetics , Female , Humans , Mice , Mice, Inbred C57BL , Viral Structural Proteins/genetics , Viral Structural Proteins/metabolism , Virus Assembly , Virus Cultivation , West Nile virus/growth & development , West Nile virus/physiology
8.
mSystems ; 1(3)2016.
Article in English | MEDLINE | ID: mdl-27822536

ABSTRACT

Several biosafety level 3 and/or 4 (BSL-3/4) pathogens are high-consequence, single-stranded RNA viruses, and their genomes, when introduced into permissive cells, are infectious. Moreover, many of these viruses are select agents (SAs), and their genomes are also considered SAs. For this reason, cDNAs and/or their derivatives must be tested to ensure the absence of infectious virus and/or viral RNA before transfer out of the BSL-3/4 and/or SA laboratory. This tremendously limits the capacity to conduct viral genomic research, particularly the application of next-generation sequencing (NGS). Here, we present a sequence-independent method to rapidly amplify viral genomic RNA while simultaneously abolishing both viral and genomic RNA infectivity across multiple single-stranded positive-sense RNA (ssRNA+) virus families. The process generates barcoded DNA amplicons that range in length from 300 to 1,000 bp, which cannot be used to rescue a virus and are stable to transport at room temperature. Our barcoding approach allows for up to 288 barcoded samples to be pooled into a single library and run across various NGS platforms without potential reconstitution of the viral genome. Our data demonstrate that this approach provides full-length genomic sequence information not only from high-titer virion preparations but it can also recover specific viral sequence from samples with limited starting material in the background of cellular RNA, and it can be used to identify pathogens from unknown samples. In summary, we describe a rapid, universal standard operating procedure that generates high-quality NGS libraries free of infectious virus and infectious viral RNA. IMPORTANCE This report establishes and validates a standard operating procedure (SOP) for select agents (SAs) and other biosafety level 3 and/or 4 (BSL-3/4) RNA viruses to rapidly generate noninfectious, barcoded cDNA amenable for next-generation sequencing (NGS). This eliminates the burden of testing all processed samples derived from high-consequence pathogens prior to transfer from high-containment laboratories to lower-containment facilities for sequencing. Our established protocol can be scaled up for high-throughput sequencing of hundreds of samples simultaneously, which can dramatically reduce the cost and effort required for NGS library construction. NGS data from this SOP can provide complete genome coverage from viral stocks and can also detect virus-specific reads from limited starting material. Our data suggest that the procedure can be implemented and easily validated by institutional biosafety committees across research laboratories.

9.
Virology ; 496: 186-193, 2016 09.
Article in English | MEDLINE | ID: mdl-27336950

ABSTRACT

West Nile virus (WNV) is a flavivirus that swept rapidly across North America in 1999, declined in prevalence, and then resurged in 2012. To date, no vaccine is available to prevent infection in the human population. Herpes simplex virus (HSV) replication-defective vaccine vectors induce a durable immunity characterized by strong antibody and CD8(+) T cell responses even in HSV-immune animals. In this study, a WNV protein expression cassette was optimized for virus-like particle (VLP) production in transfection studies, and the cassette was recombined into an HSV-1 d106-WNV virus vector, which produced extracellular VLPs, as confirmed by immunoelectron microscopy. Immunization of mice with the d106-WNV recombinant vector elicited a specific anti-WNV IgG response. This study highlights the flavivirus coding sequences needed for efficient assembly of virus-like particles. This information will facilitate generation of additional vaccine vectors against other flaviviruses including the recently emerged Zika virus.


Subject(s)
Genetic Vectors/genetics , Herpesvirus 1, Human/genetics , Vaccines, Virus-Like Particle/genetics , Viral Structural Proteins/genetics , West Nile virus/genetics , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Capsid/immunology , Capsid/ultrastructure , Cell Line , Gene Order , Humans , Immunization , Mice , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/ultrastructure , Viral Structural Proteins/chemistry , Viral Structural Proteins/immunology , West Nile virus/immunology
10.
J Virol ; 90(1): 292-9, 2016 01 01.
Article in English | MEDLINE | ID: mdl-26468544

ABSTRACT

UNLABELLED: The arthropod-borne West Nile virus (WNV) emerged in New York State in 1999 and quickly spread throughout the United States. Transmission is maintained in an enzootic cycle in which infected mosquitoes transmit the virus to susceptible hosts during probing and feeding. Arthropod-derived components within the viral inoculum are increasingly acknowledged to play a role in infection of vertebrate hosts. We previously showed that Culex tarsalis mosquito saliva and salivary gland extract (SGE) enhance the in vivo replication of WNV. Here, we characterized the effective dose, timing, and proximity of saliva and SGE administration necessary for enhancement of WNV viremia using a mouse model. Mosquito saliva and SGE enhanced viremia in a dose-dependent manner, and a single mosquito bite or as little as 0.01 µg of SGE was effective at enhancing viremia, suggesting a potent active salivary factor. Viremia was enhanced when SGE was injected in the same location as virus inoculation from 24 h before virus inoculation through 12 h after virus inoculation. These results were confirmed with mosquito saliva deposited by uninfected mosquitoes. When salivary treatment and virus inoculation were spatially separated, viremia was not enhanced. In summary, the effects of mosquito saliva and SGE were potent, long lasting, and localized, and these studies have implications for virus transmission in nature, where vertebrate hosts are fed upon by both infected and uninfected mosquitoes over time. Furthermore, our model provides a robust system to identify the salivary factor(s) responsible for enhancement of WNV replication. IMPORTANCE: Mosquito-borne viruses are a significant class of agents causing emerging infectious diseases. WNV has caused over 18,000 cases of neuroinvasive disease in the United States since its emergence. We have shown that Culex tarsalis mosquito saliva and SGE enhance the replication of WNV. We now demonstrate that saliva and SGE have potent, long-lasting, and localized effects. Our model provides a robust system to identify the salivary factor(s) and characterize the mechanism responsible for enhancement of WNV replication. These studies could lead to the identification of novel prophylactic or treatment options useful in limiting the spread of WNV, other mosquito-borne viruses, and the diseases that they cause.


Subject(s)
Culex/physiology , Saliva/metabolism , Tissue Extracts/metabolism , West Nile Fever/transmission , West Nile Fever/virology , West Nile virus/growth & development , Animals , Disease Models, Animal , Female , Mice, Inbred C57BL , Viral Load , Viremia , West Nile virus/isolation & purification
11.
Virol J ; 10: 242, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-23876037

ABSTRACT

BACKGROUND: The flaviviral nonstructural protein 5 (NS5) is a phosphoprotein, though the precise identities and roles of many specific phosphorylations remain unknown. Protein kinase G (PKG), a cGMP-dependent protein kinase, has previously been shown to phosphorylate dengue virus NS5. METHODS: We used mass spectrometry to specifically identify NS5 phosphosites. Co-immunoprecipitation assays were used to study protein-protein interactions. Effects on viral replication were measured via replicon system and plaque assay titering. RESULTS: We identified multiple sites in West Nile virus (WNV) NS5 that are phosphorylated during a WNV infection, and showed that the N-terminal methyltransferase domain of WNV NS5 can be specifically phosphorylated by PKG in vitro. Expressing PKG in cell culture led to an enhancement of WNV viral production. We hypothesized this effect on replication could be caused by factors beyond the specific phosphorylations of NS5. Here we show for the first time that PKG is also able to stably interact with a viral substrate, WNV NS5, in cell culture and in vitro. While the mosquito-borne WNV NS5 interacted with PKG, tick-borne Langat virus NS5 did not. The methyltransferase domain of NS5 is able to mediate the interaction between NS5 and PKG, and mutating positive residues in the αE region of the methyltransferase interrupts the interaction. These same mutations completely inhibited WNV replication. CONCLUSIONS: PKG is not required for WNV replication, but does make a stable interaction with NS5. While the consequence of the NS5:PKG interaction when it occurs is unclear, mutational data demonstrates that this interaction occurs in a region of NS5 that is otherwise necessary for replication. Overall, the results identify an interaction between virus and a cellular kinase and suggest a role for a host kinase in enhancing flaviviral replication.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/metabolism , Host-Pathogen Interactions , Methyltransferases/metabolism , Viral Nonstructural Proteins/metabolism , West Nile virus/physiology , Animals , Cell Line , DNA Mutational Analysis , Humans , Mutant Proteins/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Processing, Post-Translational
12.
PLoS One ; 6(10): e26697, 2011.
Article in English | MEDLINE | ID: mdl-22039536

ABSTRACT

Many viruses within the Flavivirus genus cause significant disease in humans; however, effective antivirals against these viruses are not currently available. We have previously shown that a thiopurine drug, 6-methylmercaptopurine riboside (6MMPr), inhibits replication of distantly related viruses within the Flaviviridae family in cell culture, including bovine viral diarrhea virus and hepatitis C virus replicon. Here we further examined the potential antiviral effect of 6MMPr on several diverse flaviviruses. In cell culture, 6MMPr inhibited virus production of yellow fever virus, dengue virus-2 (DENV-2) and West Nile virus (WNV) in a dose-dependent manner, and DENV-2 was significantly more sensitive to 6MMPr treatment than WNV. We then explored the use of 6MMPr as an antiviral against WNV in an immunocompetent mouse model. Once a day treatment of mice with 0.5 mg 6MMPr was just below the toxic dose in our mouse model, and this dose was used in subsequent studies. Mice were treated with 6MMPr immediately after subcutaneous inoculation with WNV for eight consecutive days. Treatment with 6MMPr exacerbated weight loss in WNV-inoculated mice and did not significantly affect mortality. We hypothesized that 6MMPr has low bioavailability in the central nervous system (CNS) and examined the effect of pre-treatment with 6MMPr on viral loads in the periphery and CNS. Pre-treatment with 6MMPr had no significant effect on viremia or viral titers in the periphery, but resulted in significantly higher viral loads in the brain, suggesting that the effect of 6MMPr is tissue-dependent. In conclusion, despite being a potent inhibitor of flaviviruses in cell culture, 6MMPr was not effective against West Nile disease in mice; however, further studies are warranted to reduce the toxicity and/or improve the bioavailability of this potential antiviral drug.


Subject(s)
Antiviral Agents/pharmacology , Methylthioinosine/pharmacology , Virus Replication/drug effects , West Nile virus/drug effects , Animals , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Mice , Vero Cells , West Nile virus/physiology
13.
J Virol ; 85(23): 12605-13, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21937657

ABSTRACT

West Nile virus (WNV) is similar to other RNA viruses in that it forms genetically complex populations within hosts. The virus is maintained in nature in mosquitoes and birds, with each host type exerting distinct influences on virus populations. We previously observed that prolonged replication in mosquitoes led to increases in WNV genetic diversity and diminished pathogenesis in mice without remarkable changes to the consensus genome sequence. We therefore sought to evaluate the relationships between individual and group phenotypes in WNV and to discover novel viral determinants of pathogenesis in mice and fitness in mosquitoes and birds. Individual plaque size variants were isolated from a genetically complex population, and mutations conferring a small-plaque and mouse-attenuated phenotype were localized to the RNA helicase domain of the NS3 protein by reverse genetics. The mutation, an Asp deletion, did not alter type I interferon production in the host but rendered mutant viruses more susceptible to interferon compared to wild type (WT) WNV. Finally, we used an in vivo fitness assay in Culex quinquefasciatus mosquitoes and chickens to determine whether the mutation in NS3 influenced fitness. The fitness of the NS3 mutant was dramatically lower in chickens and moderately lower in mosquitoes, indicating that RNA helicase is a major fitness determinant of WNV and that the effect on fitness is host specific. Overall, this work highlights the complex relationships that exist between individual and group phenotypes in RNA viruses and identifies RNA helicase as an attenuation and fitness determinant in WNV.


Subject(s)
Chickens/virology , Culicidae/virology , Genome, Viral , West Nile Fever/pathology , West Nile Fever/parasitology , West Nile virus/genetics , West Nile virus/pathogenicity , Animals , Cells, Cultured , Chickens/genetics , Chlorocebus aethiops , Cricetinae , Culicidae/genetics , Culicidae/pathogenicity , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/virology , Genetic Variation , Interferons/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/virology , Mice , Mice, Inbred C3H , Mutation/genetics , RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Viral/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication , West Nile Fever/virology
14.
J Virol ; 85(10): 5197-201, 2011 May.
Article in English | MEDLINE | ID: mdl-21367890

ABSTRACT

West Nile virus (WNV) replicates in the skin; however, cell targets in the skin have not been identified. In the current studies, WNV infected the epidermis and adnexal glands of mouse skin, and the epidermal cells were identified as keratinocytes by double labeling for WNV antigen and keratin 10. Inoculation of mice with WNV replicon particles resulted in high levels of replication in the skin, suggesting that keratinocytes are an initial target of WNV. In addition, primary keratinocytes produced infectious virus in vitro. In conclusion, keratinocytes are cell targets of WNV in vivo and may play an important role in pathogenesis.


Subject(s)
Keratinocytes/virology , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/pathogenicity , Animals , Female , Immunohistochemistry , Keratin-10/analysis , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microscopy , Skin/pathology , Skin/virology
15.
BMC Immunol ; 12: 6, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21251256

ABSTRACT

BACKGROUND: West Nile virus (WNV) persists in humans and several animal models. We previously demonstrated that WNV persists in the central nervous system (CNS) of mice for up to 6 months post-inoculation. We hypothesized that the CNS immune response is ineffective in clearing the virus. RESULTS: Immunocompetent, adult mice were inoculated subcutaneously with WNV, and the CNS immune response was examined at 1, 2, 4, 8, 12 and 16 weeks post-inoculation (wpi). Characterization of lymphocyte phenotypes in the CNS revealed elevation of CD19+ B cells for 4 wpi, CD138 plasma cells at 12 wpi, and CD4+ and CD8+ T cells for at least 12 wpi. T cells recruited to the brain were activated, and regulatory T cells (Tregs) were present for at least 12 wpi. WNV-specific antibody secreting cells were detected in the brain from 2 to 16 wpi, and virus-specific CD8+ T cells directed against an immunodominant WNV epitope were detected in the brain from 1 to 16 wpi. Furthermore, these WNV-specific immune responses occurred in mice with and without acute clinical disease. CONCLUSIONS: Virus-specific immune cells persist in the CNS of mice after WNV infection for up to 16 wpi.


Subject(s)
Central Nervous System/immunology , Central Nervous System/virology , Immunity/immunology , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/immunology , Animals , Antibodies, Viral/immunology , Antibody Specificity/immunology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Brain/immunology , Brain/virology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Lymphocyte Activation/immunology , Mice , Phenotype , Species Specificity , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology
16.
J Virol ; 85(4): 1517-27, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21147918

ABSTRACT

West Nile virus (WNV) is transmitted to vertebrate hosts primarily by infected Culex mosquitoes. Transmission of arboviruses by the bite of infected mosquitoes can potentiate infection in hosts compared to viral infection by needle inoculation. Here we examined the effect of mosquito transmission on WNV infection and systematically investigated multiple factors that differ between mosquito infection and needle inoculation of WNV. We found that mice infected with WNV through the bite of a single infected Culex tarsalis mosquito exhibited 5- to 10-fold-higher viremia and tissue titers at 24 and 48 h postinoculation and faster neuroinvasion than mice given a median mosquito-inoculated dose of WNV (10(5) PFU) by needle. Mosquito-induced enhancement was not due to differences in inoculation location, because additional intravenous inoculation of WNV did not enhance viremia or tissue titers. Inoculation of WNV into a location where uninfected mosquitoes had fed resulted in enhanced viremia and tissue titers in mice similar to those in mice infected by a single infected mosquito bite, suggesting that differences in where virus is deposited in the skin and in the virus particle itself were not responsible for the enhanced early infection in mosquito-infected mice. In addition, inoculation of mice with WNV mixed with salivary gland extract (SGE) led to higher viremia, demonstrating that mosquito saliva is the major cause of mosquito-induced enhancement. Enhanced viremia was not observed when SGE was inoculated at a distal site, suggesting that SGE enhances WNV replication by exerting a local effect. Furthermore, enhancement of WNV infection still occurred in mice with antibodies against mosquito saliva. In conclusion, saliva from C. tarsalis is responsible for enhancement of early WNV infection in vertebrate hosts.


Subject(s)
Culex/virology , Insect Vectors/virology , Saliva/virology , West Nile Fever/pathology , West Nile virus/pathogenicity , Animals , Bites and Stings/virology , Cell Line , Cells, Cultured , Chlorocebus aethiops , Female , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Vero Cells , Virus Replication , West Nile Fever/transmission , West Nile Fever/virology
17.
PLoS One ; 5(5): e10649, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20498839

ABSTRACT

Most acute infections with RNA viruses are transient and subsequently cleared from the host. Recent evidence, however, suggests that the RNA virus, West Nile virus (WNV), not only causes acute disease, but can persist long term in humans and animal models. Our goal in this study was to develop a mouse model of WNV persistence. We inoculated immunocompetent mice subcutaneously (s.c.) with WNV and examined their tissues for infectious virus and WNV RNA for 16 months (mo) post-inoculation (p.i.). Infectious WNV persisted for 1 mo p.i. in all mice and for 4 mo p.i. in 12% of mice, and WNV RNA persisted for up to 6 mo p.i. in 12% of mice. The frequency of persistence was tissue dependent and was in the following order: skin, spinal cord, brain, lymphoid tissues, kidney, and heart. Viral persistence occurred in the face of a robust antibody response and in the presence of inflammation in the brain. Furthermore, persistence in the central nervous system (CNS) and encephalitis were observed even in mice with subclinical infections. Mice were treated at 1 mo p.i. with cyclophosphamide, and active viral replication resulted, suggesting that lymphocytes are functional during viral persistence. In summary, WNV persisted in the CNS and periphery of mice for up to 6 mo p.i. in mice with subclinical infections. These results have implications for WNV-infected humans. In particular, immunosuppressed patients, organ transplantation, and long term sequelae may be impacted by WNV persistence.


Subject(s)
Central Nervous System/virology , West Nile virus/physiology , Animals , Antibody Formation/drug effects , Antibody Specificity/drug effects , Central Nervous System/pathology , Cyclophosphamide/pharmacology , Immunosuppression Therapy , Longevity/drug effects , Mice , Organ Specificity/drug effects , RNA, Viral/genetics , RNA, Viral/isolation & purification , Recurrence , West Nile Fever/immunology , West Nile Fever/pathology , West Nile Fever/virology , West Nile virus/drug effects , West Nile virus/genetics
18.
Virology ; 400(1): 93-103, 2010 Apr 25.
Article in English | MEDLINE | ID: mdl-20167345

ABSTRACT

West Nile virus (WNV) is a mosquito-borne pathogen. During replication, WNV acquires different carbohydrates and lipid membranes, depending on its mosquito or vertebrate hosts. Consequently, WNV derived from mosquito and vertebrate cell lines differ in their infectivity for dendritic cells (DCs) and induction of type I interferon (IFN-alpha/beta) in vitro. We evaluated the pathogenesis of WNV derived from mosquito (WNV(C6/36)) and vertebrate (WNV(BHK)) cell lines in mice. The tissue tropism, infectivity, clinical disease, and mortality did not differ for mice inoculated with WNV(C6/36) or WNV(BHK), and there were only minor differences in viral load and serum levels of IFN-alpha/beta. The replication kinetics of WNV(C6/36) and WNV(BHK) were equivalent in primary DCs and skin cells although primary DCs were more susceptible to WNV(C6/36) infection than to WNV(BHK) infection, suggesting that less virus is produced per infected cell for WNV(C6/36). In conclusion, viral source has minimal effect on WNV pathogenesis in vivo.


Subject(s)
West Nile virus/pathogenicity , Aedes , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Host-Pathogen Interactions , Interferon Type I/biosynthesis , Mice , Mice, Inbred C57BL , Species Specificity , Vero Cells , Virus Cultivation , Virus Replication , West Nile Fever/immunology , West Nile Fever/virology , West Nile virus/isolation & purification , West Nile virus/physiology
19.
Proc Natl Acad Sci U S A ; 106(48): 20435-9, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19918064

ABSTRACT

Dengue virus (DENV), a mosquito-borne flavivirus, is a major public health threat. The virus poses risk to 2.5 billion people worldwide and causes 50 to 100 million human infections each year. Neither a vaccine nor an antiviral therapy is currently available for prevention and treatment of DENV infection. Here, we report a previously undescribed adenosine analog, NITD008, that potently inhibits DENV both in vitro and in vivo. In addition to the 4 serotypes of DENV, NITD008 inhibits other flaviviruses, including West Nile virus, yellow fever virus, and Powassan virus. The compound also suppresses hepatitis C virus, but it does not inhibit nonflaviviruses, such as Western equine encephalitis virus and vesicular stomatitis virus. A triphosphate form of NITD008 directly inhibits the RNA-dependent RNA polymerase activity of DENV, indicating that the compound functions as a chain terminator during viral RNA synthesis. NITD008 has good in vivo pharmacokinetic properties and is biologically available through oral administration. Treatment of DENV-infected mice with NITD008 suppressed peak viremia, reduced cytokine elevation, and completely prevented the infected mice from death. No observed adverse effect level (NOAEL) was achieved when rats were orally dosed with NITD008 at 50 mg/kg daily for 1 week. However, NOAEL could not be accomplished when rats and dogs were dosed daily for 2 weeks. Nevertheless, our results have proved the concept that a nucleoside inhibitor could be developed for potential treatment of flavivirus infections.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/metabolism , Dengue/drug therapy , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Viremia/drug therapy , Adenosine/chemistry , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Male , Mice , Molecular Structure , No-Observed-Adverse-Effect Level , Rats , Vero Cells
20.
Diagn Microbiol Infect Dis ; 65(2): 150-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19748425

ABSTRACT

A duplex TaqMan real-time reverse transcriptase polymerase chain reaction (PCR) assay was developed for the detection of California (CAL) serogroup viruses and Cache Valley virus (CVV), for use in human surveillance. The targets selected for the assay were the sequences encoding the nucleocapsid protein of CAL and the G1 glycoprotein of CVV. Conserved regions were selected by aligning genetic sequences from various strains available in the GenBank database. Primers and probes were selected in conserved regions. The assay sensitivity was 75 gene copies (gc)/reaction for CAL serogroup viruses and 30 gc/reaction for CVV. The performance of the assay was linear over at least 6 log(10) gc. The assay was specific, given that it did not cross-react with a variety of pathogens. It did, however, detect 11 viruses within the CAL serogroup and 12 CVV isolates. The use of an internal control ensured that possible inefficiency in nucleic acid extraction or PCR inhibition would be detected.


Subject(s)
Bunyamwera virus/isolation & purification , Bunyaviridae Infections/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/methods , Bunyamwera virus/genetics , Conserved Sequence , DNA Primers/genetics , Humans , Nucleocapsid Proteins/genetics , Reference Standards , Reverse Transcriptase Polymerase Chain Reaction/standards , Sensitivity and Specificity , Sequence Alignment , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL