Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
1.
Immunity ; 43(6): 1112-24, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26680205

ABSTRACT

CD8(+) T cells and NK cells protect from viral infections by killing virally infected cells and secreting interferon-γ. Several inhibitory receptors limit the magnitude and duration of these anti-viral responses. NKG2A, which is encoded by Klrc1, is a lectin-like inhibitory receptor that is expressed as a heterodimer with CD94 on NK cells and activated CD8(+) T cells. Previous studies on the impact of CD94/NKG2A heterodimers on anti-viral responses have yielded contrasting results and the in vivo function of NKG2A remains unclear. Here, we generated Klrc1(-/-) mice and found that NKG2A is selectively required for resistance to ectromelia virus (ECTV). NKG2A functions intrinsically within ECTV-specific CD8(+) T cells to limit excessive activation, prevent apoptosis, and preserve the specific CD8(+) T cell response. Thus, although inhibitory receptors often cause T cell exhaustion and viral spreading during chronic viral infections, NKG2A optimizes CD8(+) T cell responses during an acute poxvirus infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , NK Cell Lectin-Like Receptor Subfamily C/immunology , Poxviridae Infections/immunology , Animals , Cytotoxicity, Immunologic/immunology , Disease Models, Animal , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
2.
Am J Physiol Regul Integr Comp Physiol ; 316(5): R525-R534, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30811246

ABSTRACT

Double-stranded (ds) RNA, both synthetic and produced during virus replication, rapidly stimulates MAPK and NF-κB signaling that results in expression of the inflammatory genes inducible nitric oxide synthase, cyclooxygenase 2, and IL-1ß by macrophages. Using biochemical and genetic approaches, we have identified the chemokine ligand-binding C-C chemokine receptor type 5 (CCR5) as a cell surface signaling receptor required for macrophage expression of inflammatory genes in response to dsRNA. Activation of macrophages by synthetic dsRNA does not require known dsRNA receptors, as poly(inosinic:cytidylic) acid [poly(I:C)] activates signaling pathways leading to expression of inflammatory genes to similar levels in wild-type and Toll-like receptor 3- or melanoma differentiation antigen 5-deficient macrophages. In contrast, macrophage activation in response to poly(I:C) is attenuated in macrophages isolated from mice lacking CCR5. These findings support a role for CCR5 as a cell surface signaling receptor that participates in activation of inflammatory genes in macrophages in response to the viral dsRNA mimetic poly(inosinic:cytidylic) acid by pathways that are distinct from classical dsRNA receptor-mediated responses.


Subject(s)
Inflammation/metabolism , Macrophage Activation/drug effects , Macrophages, Peritoneal/drug effects , Poly I-C/pharmacology , Receptors, CCR5/agonists , Signal Transduction/drug effects , Animals , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Inflammation/genetics , Inflammation/immunology , Interferon-Induced Helicase, IFIH1/deficiency , Interferon-Induced Helicase, IFIH1/genetics , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
3.
J Biol Chem ; 291(1): 447-61, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26546677

ABSTRACT

Exogenous cytokine therapy can induce systemic toxicity, which might be prevented by activating endogenously produced cytokines in local cell niches. Here we developed antibody-based activators of cytokine signaling (AcCS), which recognize cytokines only when they are bound to their cell surface receptors. AcCS were developed for type I interferons (IFNs), which induce cellular activities by binding to cell surface receptors IFNAR1 and IFNAR2. As a potential alternative to exogenous IFN therapy, AcCS were shown to potentiate the biological activities of natural IFNs by ∼100-fold. Biochemical and structural characterization demonstrates that the AcCS stabilize the IFN-IFNAR2 binary complex by recognizing an IFN-induced conformational change in IFNAR2. Using IFN mutants that disrupt IFNAR1 binding, AcCS were able to enhance IFN antiviral potency without activating antiproliferative responses. This suggests AcCS can be used to manipulate cytokine signaling for basic science and possibly for therapeutic applications.


Subject(s)
Cytokines/immunology , Immunoglobulin Fragments/immunology , Receptors, Cytokine/immunology , Signal Transduction , Antiviral Agents/chemistry , Binding Sites , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation , Humans , Immunoglobulin Fragments/pharmacology , Interferon-alpha/pharmacology , Kinetics , Mutation/genetics , Phosphorylation , Protein Conformation , Receptor, Interferon alpha-beta/chemistry , Receptor, Interferon alpha-beta/metabolism , Reproducibility of Results , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
4.
PLoS Pathog ; 11(6): e1004941, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26107264

ABSTRACT

Naïve anti-viral CD8+ T cells (TCD8+) are activated by the presence of peptide-MHC Class I complexes (pMHC-I) on the surface of professional antigen presenting cells (pAPC). Increasing the number of pMHC-I in vivo can increase the number of responding TCD8+. Antigen can be presented directly or indirectly (cross presentation) from virus-infected and uninfected cells, respectively. Here we determined the relative importance of these two antigen presenting pathways in mousepox, a natural disease of the mouse caused by the poxvirus, ectromelia (ECTV). We demonstrated that ECTV infected several pAPC types (macrophages, B cells, and dendritic cells (DC), including DC subsets), which directly presented pMHC-I to naïve TCD8+ with similar efficiencies in vitro. We also provided evidence that these same cell-types presented antigen in vivo, as they form contacts with antigen-specific TCD8+. Importantly, the number of pMHC-I on infected pAPC (direct presentation) vastly outnumbered those on uninfected cells (cross presentation), where presentation only occurred in a specialized subset of DC. In addition, prior maturation of DC failed to enhance antigen presentation, but markedly inhibited ECTV infection of DC. These results suggest that direct antigen presentation is the dominant pathway in mice during mousepox. In a broader context, these findings indicate that if a virus infects a pAPC then the presentation by that cell is likely to dominate over cross presentation as the most effective mode of generating large quantities of pMHC-I is on the surface of pAPC that endogenously express antigens. Recent trends in vaccine design have focused upon the introduction of exogenous antigens into the MHC Class I processing pathway (cross presentation) in specific pAPC populations. However, use of a pantropic viral vector that targets pAPC to express antigen endogenously likely represents a more effective vaccine strategy than the targeting of exogenous antigen to a limiting pAPC subpopulation.


Subject(s)
Antigen Presentation/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Animals , Cross-Priming/immunology , Dendritic Cells/immunology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Peptides/immunology , Peptides/metabolism , Phenotype
5.
PLoS Pathog ; 11(3): e1004770, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25822986

ABSTRACT

Imatinib mesylate (Gleevec) inhibits Abl1, c-Kit, and related protein tyrosine kinases (PTKs) and serves as a therapeutic for chronic myelogenous leukemia and gastrointestinal stromal tumors. Imatinib also has efficacy against various pathogens, including pathogenic mycobacteria, where it decreases bacterial load in mice, albeit at doses below those used for treating cancer. We report that imatinib at such low doses unexpectedly induces differentiation of hematopoietic stem cells and progenitors in the bone marrow, augments myelopoiesis but not lymphopoiesis, and increases numbers of myeloid cells in blood and spleen. Whereas progenitor differentiation relies on partial inhibition of c-Kit by imatinib, lineage commitment depends upon inhibition of other PTKs. Thus, imatinib mimics "emergency hematopoiesis," a physiological innate immune response to infection. Increasing neutrophil numbers by adoptive transfer sufficed to reduce mycobacterial load, and imatinib reduced bacterial load of Franciscella spp., which do not utilize imatinib-sensitive PTKs for pathogenesis. Thus, potentiation of the immune response by imatinib at low doses may facilitate clearance of diverse microbial pathogens.


Subject(s)
Cell Differentiation/drug effects , Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Imatinib Mesylate/pharmacology , Myelopoiesis/drug effects , Neutrophils/immunology , Animals , Cell Differentiation/immunology , Leukocyte Count , Mice , Myelopoiesis/immunology
6.
PLoS Pathog ; 10(8): e1004326, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25122471

ABSTRACT

Poxviruses contain large dsDNA genomes encoding numerous open reading frames that manipulate cellular signalling pathways and interfere with the host immune response. The NF-κB signalling cascade is an important mediator of innate immunity and inflammation, and is tightly regulated by ubiquitination at several key points. A critical step in NF-κB activation is the ubiquitination and degradation of the inhibitor of kappaB (IκBα), by the cellular SCFß-TRCP ubiquitin ligase complex. We show here that upon stimulation with TNFα or IL-1ß, Orthopoxvirus-infected cells displayed an accumulation of phosphorylated IκBα, indicating that NF-κB activation was inhibited during poxvirus infection. Ectromelia virus is the causative agent of lethal mousepox, a natural disease that is fatal in mice. Previously, we identified a family of four ectromelia virus genes (EVM002, EVM005, EVM154 and EVM165) that contain N-terminal ankyrin repeats and C-terminal F-box domains that interact with the cellular SCF ubiquitin ligase complex. Since degradation of IκBα is catalyzed by the SCFß-TRCP ubiquitin ligase, we investigated the role of the ectromelia virus ankyrin/F-box protein, EVM005, in the regulation of NF-κB. Expression of Flag-EVM005 inhibited both TNFα- and IL-1ß-stimulated IκBα degradation and p65 nuclear translocation. Inhibition of the NF-κB pathway by EVM005 was dependent on the F-box domain, and interaction with the SCF complex. Additionally, ectromelia virus devoid of EVM005 was shown to inhibit NF-κB activation, despite lacking the EVM005 open reading frame. Finally, ectromelia virus devoid of EVM005 was attenuated in both A/NCR and C57BL/6 mouse models, indicating that EVM005 is required for virulence and immune regulation in vivo.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/metabolism , NF-kappa B/metabolism , Viral Proteins/metabolism , Animals , Ectromelia virus/immunology , Ectromelia virus/metabolism , Ectromelia, Infectious/immunology , Flow Cytometry , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , NF-kappa B/immunology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Viral Proteins/immunology , Virulence/physiology
7.
Nature ; 468(7322): 452-6, 2010 Nov 18.
Article in English | MEDLINE | ID: mdl-21085181

ABSTRACT

Cellular messenger RNA (mRNA) of higher eukaryotes and many viral RNAs are methylated at the N-7 and 2'-O positions of the 5' guanosine cap by specific nuclear and cytoplasmic methyltransferases (MTases), respectively. Whereas N-7 methylation is essential for RNA translation and stability, the function of 2'-O methylation has remained uncertain since its discovery 35 years ago. Here we show that a West Nile virus (WNV) mutant (E218A) that lacks 2'-O MTase activity was attenuated in wild-type primary cells and mice but was pathogenic in the absence of type I interferon (IFN) signalling. 2'-O methylation of viral RNA did not affect IFN induction in WNV-infected fibroblasts but instead modulated the antiviral effects of IFN-induced proteins with tetratricopeptide repeats (IFIT), which are interferon-stimulated genes (ISGs) implicated in regulation of protein translation. Poxvirus and coronavirus mutants that lacked 2'-O MTase activity similarly showed enhanced sensitivity to the antiviral actions of IFN and, specifically, IFIT proteins. Our results demonstrate that the 2'-O methylation of the 5' cap of viral RNA functions to subvert innate host antiviral responses through escape of IFIT-mediated suppression, and suggest an evolutionary explanation for 2'-O methylation of cellular mRNA: to distinguish self from non-self RNA. Differential methylation of cytoplasmic RNA probably serves as an example for pattern recognition and restriction of propagation of foreign viral RNA in host cells.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation/immunology , Immunity, Innate/immunology , Interferons/immunology , Proteins/metabolism , RNA Caps/metabolism , RNA, Viral/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Cells, Cultured , Coronavirus/enzymology , Coronavirus/genetics , Coronavirus/immunology , Coronavirus/physiology , Fibroblasts , Gene Expression Regulation/genetics , Humans , Immunity, Innate/genetics , Interferons/deficiency , Interferons/genetics , Methylation , Methyltransferases/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Models, Immunological , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Poxviridae/enzymology , Poxviridae/genetics , Poxviridae/immunology , Poxviridae/physiology , Protein Biosynthesis/immunology , Proteins/genetics , RNA Caps/genetics , RNA Caps/immunology , RNA, Viral/genetics , RNA, Viral/immunology , RNA-Binding Proteins , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Survival Rate , Virus Replication , West Nile virus/enzymology , West Nile virus/genetics , West Nile virus/immunology , West Nile virus/physiology
8.
Antimicrob Agents Chemother ; 58(12): 7171-81, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25224011

ABSTRACT

Adenovirus infections of immunocompromised patients can develop into deadly multiorgan or systemic disease. The virus is especially threatening for pediatric allogeneic hematopoietic stem cell transplant recipients; according to some studies, 10% or more of these patients succumb to disease resulting from adenovirus infection. At present, there is no drug approved for the treatment or prevention of adenovirus infections. Compounds that are approved to treat other virus infections are used off-label to combat adenovirus, but only anecdotal evidence of the efficacy of these drugs exists. Ganciclovir, a drug approved for the treatment of herpesvirus infection, was previously reported to be effective against human adenoviruses in vitro. To model adenovirus infections in immunocompromised humans, we examined ganciclovir's efficacy in immunosuppressed Syrian hamsters intravenously infected with type 5 human adenovirus (Ad5). This animal model is permissive for Ad5 replication, and the animals develop symptoms similar to those seen in humans. We demonstrate that ganciclovir suppresses Ad5 replication in the liver of infected hamsters and that it mitigates the consequences of Ad5 infections in these animals when administered prophylactically or therapeutically. We show that ganciclovir inhibits Ad5 DNA synthesis and late gene expression. The mechanism of action for the drug is not clear; preliminary data suggest that it exerts its antiadenoviral effect by directly inhibiting the adenoviral DNA polymerase. While more extensive studies are required, we believe that ganciclovir is a promising drug candidate to treat adenovirus infections. Brincidofovir, a drug with proven activity against Ad5, was used as a positive control in the prophylactic experiment.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Ganciclovir/pharmacology , Immunocompromised Host , Viral Proteins/antagonists & inhibitors , Adenoviridae Infections/immunology , Adenoviridae Infections/mortality , Adenoviridae Infections/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/growth & development , Adenoviruses, Human/pathogenicity , Animals , Body Weight/drug effects , Cell Line, Tumor , Cytosine/analogs & derivatives , Cytosine/pharmacology , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Female , Gene Expression , Humans , Male , Mesocricetus , Organophosphonates/pharmacology , Survival Analysis , Transaminases/blood , Viral Load/drug effects , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
9.
J Virol ; 87(9): 4846-60, 2013 May.
Article in English | MEDLINE | ID: mdl-23408632

ABSTRACT

Rift Valley fever virus (RVFV) is an emerging RNA virus with devastating economic and social consequences. Clinically, RVFV induces a gamut of symptoms ranging from febrile illness to retinitis, hepatic necrosis, hemorrhagic fever, and death. It is known that type I interferon (IFN) responses can be protective against severe pathology; however, it is unknown which innate immune receptor pathways are crucial for mounting this response. Using both in vitro assays and in vivo mucosal mouse challenge, we demonstrate here that RNA helicases are critical for IFN production by immune cells and that signaling through the helicase adaptor molecule MAVS (mitochondrial antiviral signaling) is protective against mortality and more subtle pathology during RVFV infection. In addition, we demonstrate that Toll-like-receptor-mediated signaling is not involved in IFN production, further emphasizing the importance of the RNA cellular helicases in type I IFN responses to RVFV.


Subject(s)
DEAD-box RNA Helicases/immunology , Interferon-beta/immunology , Mucous Membrane/virology , Rift Valley Fever/enzymology , Rift Valley Fever/immunology , Rift Valley fever virus/physiology , Animals , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Humans , Interferon-beta/genetics , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/genetics , Mucous Membrane/immunology , Rift Valley Fever/prevention & control , Rift Valley Fever/virology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
10.
Virus Genes ; 48(1): 38-47, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24078045

ABSTRACT

Poxviruses are dsDNA viruses with large genomes. Many genes in the genome remain uncharacterized, and recent studies have demonstrated that the poxvirus transcriptome includes numerous so-called anomalous transcripts not associated with open reading frames. Here, we characterize the expression and role of an apparently non-coding RNA in orthopoxviruses, which we call viral hairpin RNA (vhRNA). Using a bioinformatics approach, we predicted expression of a transcript not associated with an open reading frame that is likely to form a stem-loop structure due to the presence of a 21 nt palindromic sequence. Expression of the transcript as early as 2 h post-infection was confirmed by northern blot and analysis of publicly available vaccinia virus infected cell transcriptomes. The transcription start site was determined by RACE PCE and transcriptome analysis, and early and late promoter sequences were identified. Finally, to test the function of the transcript we generated an ectromelia virus knockout, which failed to form plaques in cell culture. The important role of the transcript in viral replication was further demonstrated using siRNA. Although the function of the transcript remains unknown, our work contributes to evidence of an increasingly complex poxvirus transcriptome, suggesting that transcripts such as vhRNA not associated with an annotated open reading frame can play an important role in viral replication.


Subject(s)
Ectromelia virus/growth & development , Ectromelia virus/genetics , Gene Expression Regulation, Viral , Gene Expression , RNA, Untranslated/biosynthesis , Viral Plaque Assay , Animals , Blotting, Northern , Cell Line , Chlorocebus aethiops , Computational Biology , Gene Knockout Techniques , Macaca mulatta , Nucleic Acid Conformation , Promoter Regions, Genetic , RNA, Untranslated/genetics , Transcription Initiation Site , Transcription, Genetic
11.
Appl Environ Microbiol ; 79(4): 1333-41, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263945

ABSTRACT

Protection of the human lung from infectious agents, allergens, and ultrafine particles is difficult with current technologies. High-efficiency particulate air (HEPA) filters remove airborne particles of >0.3 µm with 99.97% efficiency, but they are expensive to maintain. Electrostatic precipitation has been used as an inexpensive approach to remove large particles from airflows, but it has a collection efficiency minimum in the submicrometer size range, allowing for a penetration window for some allergens and ultrafine particles. Incorporating soft X-ray irradiation as an in situ component of the electrostatic precipitation process greatly improves capture efficiency of ultrafine particles. Here we demonstrate the removal and inactivation capabilities of soft-X-ray-enhanced electrostatic precipitation technology targeting infectious agents (Bacillus anthracis, Mycobacterium bovis BCG, and poxviruses), allergens, and ultrafine particles. Incorporation of in situ soft X-ray irradiation at low-intensity corona conditions resulted in (i) 2-fold to 9-fold increase in capture efficiency of 200- to 600-nm particles and (ii) a considerable delay in the mean day of death as well as lower overall mortality rates in ectromelia virus (ECTV) cohorts. At the high-intensity corona conditions, nearly complete protection from viral and bacterial respiratory infection was afforded to the murine models for all biological agents tested. When optimized for combined efficient particle removal with limited ozone production, this technology could be incorporated into stand-alone indoor air cleaners or scaled for installation in aircraft cabin, office, and residential heating, ventilating, and air-conditioning (HVAC) systems.


Subject(s)
Air Microbiology , Allergens/radiation effects , Bacteria/radiation effects , Chemical Precipitation , Particulate Matter/radiation effects , Viruses/radiation effects , X-Rays , Allergens/chemistry , Animals , Bacteria/chemistry , Disease Models, Animal , Mice , Particulate Matter/chemistry , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/prevention & control , Respiratory Tract Infections/virology , Static Electricity , Viruses/chemistry
12.
J Virol ; 85(21): 11170-82, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21849445

ABSTRACT

Ectromelia virus (ECTV) is a natural pathogen of mice that causes mousepox, and many of its genes have been implicated in the modulation of host immune responses. Serine protease inhibitor 2 (SPI-2) is one of these putative ECTV host response modifier proteins. SPI-2 is conserved across orthopoxviruses, but results defining its mechanism of action and in vivo function are lacking or contradictory. We studied the role of SPI-2 in mousepox by deleting the SPI-2 gene or its serine protease inhibitor reactive site. We found that SPI-2 does not affect viral replication or cell-intrinsic apoptosis pathways, since mutant viruses replicate in vitro as efficiently as wild-type virus. However, in the absence of SPI-2 protein, ECTV is attenuated in mousepox-susceptible mice, resulting in lower viral loads in the liver, decreased spleen pathology, and substantially improved host survival. This attenuation correlates with more effective immune responses in the absence of SPI-2, including an earlier serum gamma interferon (IFN-γ) response, raised serum interleukin 18 (IL-18), increased numbers of granzyme B(+) CD8(+) T cells, and, most notably, increased numbers and activation of NK cells. Both virus attenuation and the improved immune responses associated with SPI-2 deletion from ECTV are lost when mice are depleted of NK cells. Consequently, SPI-2 renders mousepox lethal in susceptible strains by preventing protective NK cell defenses.


Subject(s)
Ectromelia virus/pathogenicity , Ectromelia, Infectious/mortality , Host-Pathogen Interactions , Killer Cells, Natural/immunology , Serpins/metabolism , Viral Proteins/metabolism , Virulence Factors/metabolism , Animals , Ectromelia virus/genetics , Ectromelia virus/immunology , Ectromelia, Infectious/virology , Gene Deletion , Interferon-gamma/metabolism , Interleukin-18/metabolism , Liver/virology , Lymphocyte Subsets/chemistry , Lymphocyte Subsets/immunology , Mice , Serpins/genetics , Spleen/pathology , Survival Analysis , Viral Load , Viral Proteins/genetics , Virus Replication
13.
Am J Pathol ; 179(6): 2941-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001348

ABSTRACT

Encephalomyocarditis virus (EMCV) is capable of stimulating inflammatory gene expression by macrophages as a result of interactions between EMCV capsid proteins and cell surface receptors. In this study, biochemical and genetic approaches identified a role for Ccr5, a chemokine receptor, in transducing the signals of EMCV infection that result in the expression of inflammatory genes in macrophages. Antibody neutralization and gene knockout strategies were used to show that the presence of Ccr5 is required for EMCV-stimulated mitogen-activated protein (MAP) kinase and nuclear factor-kappa B (NF-κB) activation, and the subsequent expression of the inflammatory gene-inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2). Ccr5 appears to participate in the early control of virus replication: EMCV mRNA accumulates to sevenfold higher levels in Ccr5-deficient mice when compared to wild-type controls. These findings support a regulatory role for Ccr5 in the antiviral response to EMCV in which this chemokine receptor participates in regulation of inflammatory gene expression in response to virus infection.


Subject(s)
Encephalomyocarditis virus/physiology , Interferon Type I/biosynthesis , Macrophages/virology , Receptors, CCR5/physiology , Signal Transduction/physiology , Animals , Antibodies, Neutralizing/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , DEAD-box RNA Helicases/physiology , Gene Expression , Interferon-Induced Helicase, IFIH1 , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Receptors, CCR5/deficiency , Toll-Like Receptor 3/physiology
14.
J Virol ; 84(18): 9128-39, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20610727

ABSTRACT

Poxviruses produce complement regulatory proteins to subvert the host's immune response. Similar to the human pathogen variola virus, ectromelia virus has a limited host range and provides a mouse model where the virus and the host's immune response have coevolved. We previously demonstrated that multiple components (C3, C4, and factor B) of the classical and alternative pathways are required to survive ectromelia virus infection. Complement's role in the innate and adaptive immune responses likely drove the evolution of a virus-encoded virulence factor that regulates complement activation. In this study, we characterized the ectromelia virus inhibitor of complement enzymes (EMICE). Recombinant EMICE regulated complement activation on the surface of CHO cells, and it protected complement-sensitive intracellular mature virions (IMV) from neutralization in vitro. It accomplished this by serving as a cofactor for the inactivation of C3b and C4b and by dissociating the catalytic domain of the classical pathway C3 convertase. Infected murine cells initiated synthesis of EMICE within 4 to 6 h postinoculation. The levels were sufficient in the supernatant to protect the IMV, upon release, from complement-mediated neutralization. EMICE on the surface of infected murine cells also reduced complement activation by the alternative pathway. In contrast, classical pathway activation by high-titer antibody overwhelmed EMICE's regulatory capacity. These results suggest that EMICE's role is early during infection when it counteracts the innate immune response. In summary, ectromelia virus produced EMICE within a few hours of an infection, and EMICE in turn decreased complement activation on IMV and infected cells.


Subject(s)
Complement Inactivating Agents/immunology , Complement System Proteins/immunology , Ectromelia virus/immunology , Immune Evasion , Viral Proteins/immunology , Virulence Factors/immunology , Animals , Cell Line , Chlorocebus aethiops , Cricetinae , Cricetulus , Ectromelia virus/pathogenicity , Mice , Mice, Inbred C57BL , Neutralization Tests
15.
J Virol ; 84(8): 3909-20, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20130052

ABSTRACT

Monkeypox virus (MPXV) is an orthopoxvirus closely related to variola, the etiological agent of smallpox. In humans, MPXV causes a disease similar to smallpox and is considered to be an emerging infectious disease. Moreover, the use of MPXV for bioterroristic/biowarfare activities is of significant concern. Available small animal models of human monkeypox have been restricted to mammals with poorly defined biologies that also have limited reagent availability. We have established a murine MPXV model utilizing the STAT1-deficient C57BL/6 mouse. Here we report that a relatively low-dose intranasal (IN) infection induces 100% mortality in the stat1(-)(/)(-) model by day 10 postinfection with high infectious titers in the livers, spleens, and lungs of moribund animals. Vaccination with modified vaccinia virus Ankara (MVA) followed by a booster vaccination is sufficient to protect against an intranasal MPXV challenge and induces an immune response more robust than that of a single vaccination. Furthermore, antiviral treatment with CMX001 (HDP-cidofovir) and ST-246 protects when administered as a regimen initiated on the day of infection. Thus, the stat1(-)(/)(-) model provides a lethal murine platform for evaluating therapeutics and for investigating the immunological and pathological responses to MPXV infection.


Subject(s)
Disease Models, Animal , Monkeypox virus/drug effects , Monkeypox virus/pathogenicity , Mpox (monkeypox)/drug therapy , Mpox (monkeypox)/prevention & control , Animals , Antiviral Agents/therapeutic use , Benzamides , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Female , Humans , Isoindoles , Liver/virology , Lung/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mpox (monkeypox)/mortality , Organophosphonates/therapeutic use , STAT1 Transcription Factor/deficiency , Smallpox Vaccine/immunology , Spleen/virology , Survival Analysis , Treatment Outcome , Vaccinia virus/immunology , Viral Load
16.
Bioorg Med Chem ; 19(9): 2950-8, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21493074

ABSTRACT

Alkoxyalkyl esters of cidofovir (CDV) are orally active agents which inhibit the replication of a variety of double stranded DNA (dsDNA) viruses including variola, vaccinia, ectromelia, herpes simplex virus, cytomegalovirus, adenovirus and others. One of these compounds, hexadecyloxypropyl-CDV (HDP-CDV, CMX001) is in clinical development for prevention and treatment of poxvirus infection, vaccination complications, and for infections caused by cytomegalovirus, adenovirus, herpesviruses and other dsDNA viruses. This class of lipid analogs is potentially prone to undergo omega oxidation of the alkyl moiety which can lead to a short chain carboxylic acid lacking antiviral activity. To address this issue, we synthesized a series of alkoxyalkyl or alkyl glycerol esters of CDV and (S)-HPMPA having modifications in the structure of the alkyl residue. Antiviral activity was assessed in cells infected with vaccinia, cowpox or ectromelia viruses. Metabolic stability was determined in S9 membrane fractions from rat, guinea pig, monkey and human liver. All compounds had substantial antiviral activity in cells infected with vaccinia, cowpox or ectromelia. Metabolic stability was lowest in monkey liver S9 incubations where rapid disappearance of HDP-CDV and HDP-(S)-HPMPA was noted. Metabolic stability in monkey preparations increased substantially when a ω-1 methyl group (15-methyl-HDP-CDV) or a terminal cyclopropyl residue (14-cyclopropyl-tetradecyloxypropyl-CDV) was present in the alkyl chain. The most stable compound was 1-O-octadecyl-2-O-benzyl-sn-glycero-3-CDV (ODBG-CDV) which was not metabolized extensively by monkey liver S9. In rat, guinea pig or human liver S9 incubations, most of the modified antiviral compounds were considerably more stable.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/chemical synthesis , Cytosine/analogs & derivatives , Organophosphonates/chemistry , Adenine/chemical synthesis , Adenine/chemistry , Adenine/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cidofovir , Cowpox virus/drug effects , Cytosine/chemical synthesis , Cytosine/chemistry , Cytosine/pharmacology , Ectromelia virus/drug effects , Esters , Guinea Pigs , Haplorhini , Humans , Liver/metabolism , Organophosphonates/chemical synthesis , Organophosphonates/pharmacology , Rats , Vaccinia virus/drug effects
17.
Proc Natl Acad Sci U S A ; 105(20): 7293-7, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18490659

ABSTRACT

Adenoviruses (Ads) cause a wide array of end-organ and disseminated diseases in severely immunosuppressed patients. For example, approximately 20% of pediatric allogeneic hematopoietic stem cell transplant recipients develop disseminated Ad infection, and the disease proves fatal in as many as 50-80% of these patients. Ad infections are a serious problem for solid-organ transplant recipients and AIDS patients as well. Unfortunately, there are no antiviral drugs approved specifically to treat these infections. A suitable animal model that is permissive for Ad replication would help in the discovery process. Here we identify an animal model to study Ad pathogenesis and the efficacy of antiviral compounds. We show that human serotype 5 Ad (Ad5) causes severe systemic disease in immunosuppressed Syrian hamsters that is similar to that seen in immunocompromised patients. We also demonstrate that hexadecyloxypropyl-cidofovir (CMX001) rescues the hamsters from a lethal challenge with Ad5. The antiviral drug provided protection both prophylactically and when given up to 2 days after i.v. exposure to Ad5. CMX001 acts by reducing Ad replication in key target organs. Thus, the immunosuppressed Syrian hamster is a powerful model to evaluate anti-Ad drugs, and its use can facilitate the entry of drugs such as CMX001 into clinical trials.


Subject(s)
Cytosine/analogs & derivatives , Immunosuppressive Agents/therapeutic use , Organophosphonates/pharmacology , Adenoviridae/metabolism , Adenoviridae Infections/metabolism , Adenoviridae Infections/therapy , Animals , Antiviral Agents/pharmacology , Cricetinae , Cytosine/pharmacology , Disease Models, Animal , Drug Screening Assays, Antitumor , Hematopoietic Stem Cells/cytology , Humans , Liver/metabolism , Mesocricetus , Models, Biological
18.
Proc Natl Acad Sci U S A ; 105(6): 1861-6, 2008 Feb 12.
Article in English | MEDLINE | ID: mdl-18252829

ABSTRACT

Ectromelia virus (ECTV) encodes an IFN-gamma-binding protein (IFN-gammaBP(ECTV)) that disrupts IFN-gamma signaling and its ability to induce an antiviral state within cells. IFN-gammaBP(ECTV) is an important virulence factor that is highly conserved (>90%) in all orthopoxviruses, including variola virus, the causative agent of smallpox. The 2.2-A crystal structure of the IFN-gammaBP(ECTV)/IFN-gamma complex reveals IFN-gammaBP(ECTV) consists of an IFN-gammaR1 ligand-binding domain and a 57-aa helix-turn-helix (HTH) motif that is structurally related to the transcription factor TFIIA. The HTH motif forms a tetramerization domain that results in an IFN-gammaBP(ECTV)/IFN-gamma complex containing four IFN-gammaBP(ECTV) chains and two IFN-gamma dimers. The structure, combined with biochemical and cell-based assays, demonstrates that IFN-gammaBP(ECTV) tetramers are required for efficient IFN-gamma antagonism.


Subject(s)
Interferon-gamma/antagonists & inhibitors , Orthopoxvirus/metabolism , Viral Proteins/metabolism , Animals , Chromatography, Affinity , Hydrogen Bonding , Interferon-gamma/metabolism , Mice , Protein Binding , Protein Conformation , Viral Proteins/chemistry , Viral Proteins/isolation & purification
19.
Antiviral Res ; 195: 105179, 2021 11.
Article in English | MEDLINE | ID: mdl-34530009

ABSTRACT

Orthopoxviruses such as variola and monkeypox viruses continue to threaten the human population. Monkeypox virus is endemic in central and western Africa and outbreaks have reached as far as the U.S. Although variola virus, the etiologic agent of smallpox, has been eradicated by a successful vaccination program, official and likely clandestine stocks of the virus exist. Moreover, studies with ectromelia virus (the etiological agent of mousepox) have revealed that IL-4 recombinant viruses are significantly more virulent than wild-type viruses even in mice treated with vaccines and/or antivirals. For these reasons, it is critical that antiviral modalities are developed to treat these viruses should outbreaks, or deliberate dissemination, occur. Currently, 2 antivirals (brincidofovir and tecovirimat) are in the U.S. stockpile allowing for emergency use of the drugs to treat smallpox. Both antivirals have advantages and disadvantages in a clinical and emergency setting. Here we report on the efficacy of a recombinant immunoglobulin (rVIG) that demonstrated efficacy against several orthopoxviruses in vitro and in vivo in both a prophylactic and therapeutic fashion. A single intraperitoneal injection of rVIG significantly protected mice when given up to 14 days before or as late as 6 days post challenge. Moreover, rVIG reduced morbidity, as measured by weight-change, as well as several previously established biomarkers of disease. In rVIG treated mice, we found that vDNA levels in blood were significantly reduced, as was ALT (a marker of liver damage) and infectious virus levels in the liver. No apparent adverse events were observed in rVIG treated mice, suggesting the immunoglobulin is well tolerated. These findings suggest that recombinant immunoglobulins could be candidates for further evaluation and possible licensure under the FDA Animal Rule.


Subject(s)
Antiviral Agents/therapeutic use , Immunoglobulins/therapeutic use , Orthopoxvirus/drug effects , Smallpox/drug therapy , Vaccinia/drug therapy , Animals , Antiviral Agents/administration & dosage , Benzamides , Cell Line , Chlorocebus aethiops , Cytosine/analogs & derivatives , Female , Humans , Isoindoles , Mice , Mice, Inbred BALB C , Organophosphonates , Smallpox/prevention & control , Smallpox/virology , Smallpox Vaccine/administration & dosage , Vaccines, DNA/administration & dosage , Vaccinia/prevention & control , Vaccinia/virology
20.
J Gen Virol ; 91(Pt 9): 2278-85, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20505008

ABSTRACT

Src family kinases (SFKs) are non-receptor tyrosine kinases that have been implicated as regulators of the inflammatory response. In this study, the role of SFK activation in the inflammatory response of macrophages to encephalomyocarditis virus (EMCV) infection was examined. Virus infection of macrophages stimulates the expression of cyclooxygenase-2 (COX-2), interleukin (IL)-1beta and inducible nitric oxide synthase (iNOS). Inhibition of SFK attenuates EMCV-induced COX-2 expression and prostaglandin E(2) production, iNOS expression and subsequent nitric oxide production, and IL-1beta expression. EMCV-induced COX-2 expression requires the activation of nuclear factor-kappaB and the mitogen-activated protein kinase p38. Consistent with these previous findings, inhibition of SFKs attenuated the phosphorylation of p38 in response to EMCV infection, suggesting that SFKs may act upstream of p38. These findings provide evidence that SFK activation plays an active role in the regulation of inflammatory gene expression by virus-infected macrophages.


Subject(s)
Cyclooxygenase 2/biosynthesis , Encephalomyocarditis virus/pathogenicity , Macrophages/metabolism , Macrophages/virology , src-Family Kinases/metabolism , Animals , Base Sequence , Cell Line , Cyclooxygenase 2/genetics , DNA Primers/genetics , Dinoprostone/biosynthesis , Enzyme Activation , Host-Pathogen Interactions , I-kappa B Proteins/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Mice , Models, Biological , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL