Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
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
4.
Antiviral Res ; 112: 38-46, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25453340

ABSTRACT

There are no drugs approved specifically to treat disseminated adenovirus (Ad) infections in humans. Cidofovir is active against Ad in cell culture, and it is used frequently in the clinic with disseminated infection in pediatric transplant patients; however, controlled clinical studies have not been conducted to prove the anti-Ad efficacy of cidofovir. Brincidofovir, a lipid-linked derivative of cidofovir, which has strong activity against Ad in cell culture and in animal models, is a promising new drug currently in clinical trials. Ribavirin, which has modest activity against some Ad types in cell culture, has been used in the clinic against disseminated Ad, but the efficacy of ribavirin is unknown. In the current study, we have examined the activity of cidofovir, brincidofovir, and ribavirin against disseminated Ad5 infection in the immunosuppressed Syrian hamster model. Hamsters are immunosuppressed by treatment with cyclophosphamide, then infected intravenously with Ad5, leading to disseminated Ad5 infection, especially in the liver. We found that cidofovir and brincidofovir have excellent activity against Ad5 pathology and replication in the liver, even when administered therapeutically starting at 3 days post-challenge with Ad5. Ribavirin did not have anti-Ad5 activity in our model. Our data support the use of cidofovir and brincidofovir in humans for the treatment of disseminated Ad infections in humans.


Subject(s)
Adenoviridae Infections/drug therapy , Adenoviridae Infections/pathology , Adenoviruses, Human/growth & development , Cytosine/analogs & derivatives , Organophosphonates/therapeutic use , Ribavirin/therapeutic use , Adenoviridae Infections/virology , Alanine Transaminase/blood , Animals , Body Weight , Cell Line , Cidofovir , Cytosine/therapeutic use , Disease Models, Animal , Female , Humans , Immunocompromised Host , Liver/pathology , Liver/virology , Mesocricetus , Survival Analysis , Treatment Outcome , Viral Load
5.
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
6.
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
7.
J Clin Cell Immunol ; 4(2)2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23956946

ABSTRACT

Type 1 diabetes results from autoimmune destruction of the insulin producing pancreatic ß-cells. The immunoproteasome, a version of the proteasome that collaborates with the 11S/PA28 activator to generate immunogenic peptides for presentation by MHC class I molecules, has long been implicated in the onset of the disease, but little is known about immunoproteasome function and regulation in pancreatic ß-cells. Interesting insight into these issues comes from a recent analysis of the immunoproteasome expressed in pancreatic ß-cells during early antiviral defenses mediated by interferon ß (IFNß), a type I IFN implicated in the induction of the diabetic state in human and animal models. Using mouse islets and the MIN6 insulinoma cell line, Freudenburg et al. found that IFNß stimulates expression of the immunoproteasome and the 11S/PA28 activator in a manner fundamentally similar to the classic immuno-inducer IFNγ, with similar timing of mRNA accumulation and decline; similar transcriptional activation mediated primarily by the IRF1 and similar mRNA and protein levels. Furthermore, neither IFNß nor IFNγ altered the expression of regular proteolytic subunits or prevented their incorporation into proteolytic cores. As a result, immunoproteasomes had stochastic combinations of immune and regular proteolytic sites, an arrangement that would likely increase the probability with which unique immunogenic peptides are produced. However, immunoproteasomes were activated by the 11S/PA28 only under conditions of ATP depletion. A mechanism that prevents the activation of immunoproteasome at high ATP levels has not been reported before and could have a major regulatory significance, as it could suppress the generation of immunogenic peptides as cell accumulate immunoproteasome and 11S/PA28, and activate antigen processing only when ATP levels drop. We discuss implications of these new findings on the link between early antiviral response and the onset of type 1 diabetes.

8.
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
9.
PLoS One ; 8(2): e52408, 2013.
Article in English | MEDLINE | ID: mdl-23383295

ABSTRACT

Autoimmune destruction of insulin producing pancreatic ß-cells is the hallmark of type I diabetes. One of the key molecules implicated in the disease onset is the immunoproteasome, a protease with multiple proteolytic sites that collaborates with the constitutive 19S and the inducible 11S (PA28) activators to produce immunogenic peptides for presentation by MHC class I molecules. Despite its importance, little is known about the function and regulation of the immunoproteasome in pancreatic ß-cells. Of special interest to immunoproteasome activation in ß-cells are the effects of IFNß, a type I IFN secreted by virus-infected cells and implicated in type I diabetes onset, compared to IFNγ, the classic immunoproteasome inducer secreted by cells of the immune system. By qPCR analysis, we show that mouse insulinoma MIN6 cells and mouse islets accumulate the immune proteolytic ß1(i), ß2(i) and ß5(i), and 11S mRNAs upon exposure to IFNß or IFNγ. Higher concentrations of IFNß than IFNγ are needed for similar expression, but in each case the expression is transient, with maximal mRNA accumulation in 12 hours, and depends primarily on Interferon Regulatory Factor 1. IFNs do not alter expression of regular proteasome genes, and in the time frame of IFNß-mediated response, the immune and regular proteolytic subunits co-exist in the 20S particles. In cell extracts with ATP, these particles have normal peptidase activities and degrade polyubiquitinated proteins with rates typical of the regular proteasome, implicating normal regulation by the 19S activator. However, ATP depletion rapidly stimulates the catalytic rates in a manner consistent with levels of the 11S activator. These findings suggest that stochastic combination of regular and immune proteolytic subunits may increase the probability with which unique immunogenic peptides are produced in pancreatic ß-cells exposed to IFNß, but primarily in cells with reduced ATP levels that stimulate the 11S participation in immunoproteasome function.


Subject(s)
Adenosine Triphosphate/metabolism , Insulin-Secreting Cells/immunology , Interferon-beta/metabolism , Muscle Proteins/metabolism , Proteasome Endopeptidase Complex/immunology , Animals , Blotting, Western , Cell Line, Tumor , Chromatography, High Pressure Liquid , DNA Primers/genetics , Immunoprecipitation , Insulin-Secreting Cells/virology , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Octoxynol , Polymerase Chain Reaction , Proteasome Endopeptidase Complex/metabolism
10.
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
11.
Adv Virol ; 2012: 320206, 2012.
Article in English | MEDLINE | ID: mdl-22754567
12.
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
13.
Virology ; 409(2): 328-37, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21071055

ABSTRACT

In 2001, Jackson et al. reported that murine IL-4 expression by a recombinant ectromelia virus caused enhanced morbidity and lethality in resistant C57BL/6 mice as well as overcame protective immune memory responses. To achieve a more thorough understanding of this phenomenon and to assess a variety of countermeasures, we constructed a series of ECTV recombinants encoding murine IL-4 under the control of promoters of different strengths and temporal regulation. We showed that the ECTV-IL-4 recombinant expressing the highest level of IL-4 was uniformly lethal for C57BL/6 mice even when previously immunized. The lethality of the ECTV-IL-4 recombinants resulted from virus-expressed IL-4 signaling through the IL-4 receptor but was not due to IL-4 toxicity. A number of treatment approaches were evaluated against the most virulent IL-4 encoding virus. The most efficacious therapy was a combination of two antiviral drugs (CMX001(®) and ST-246(®)) that have different mechanisms of action.


Subject(s)
Ectromelia virus/immunology , Ectromelia virus/pathogenicity , Interleukin-4/biosynthesis , Interleukin-4/immunology , Animals , Antiviral Agents/therapeutic use , Benzamides/therapeutic use , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Ectromelia virus/genetics , Ectromelia, Infectious/drug therapy , Ectromelia, Infectious/virology , Female , Gene Expression Regulation , Interleukin-4/genetics , Isoindoles/therapeutic use , Mice , Mice, Inbred C57BL , Organophosphonates/therapeutic use , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Survival Analysis , Treatment Outcome
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 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
16.
J Innate Immun ; 2(1): 77-86, 2010.
Article in English | MEDLINE | ID: mdl-20375625

ABSTRACT

Virus infection of macrophages stimulates the expression of proinflammatory and antiviral genes interleukin-1 (IL-1), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). In this study, we show that phosphatidylinositol 3-kinase (PI3K) is required for the inflammatory response of macrophages to virus infection. When macrophages are infected with encephalomyocarditis virus (EMCV) there is a rapid and transient activation of PI3K and phosphorylation of its downstream target Akt. Inhibitors of PI3K attenuate EMCV- and double-stranded RNA-induced iNOS, COX-2 and IL-1 beta expression in RAW264.7 cells and mouse peritoneal macrophages. The attenuation of inflammatory gene expression in response to PI3K inhibition correlates with the induction of macrophage apoptosis. The morphology of macrophages shifts from activation in response to EMCV infection to apoptosis in the cells treated with PI3K inhibitors and EMCV. These morphological changes are accompanied by the activation of caspase-3. These findings suggest that PI3K plays a central role in the regulation of macrophage responses to EMCV infection. When PI3K is activated, it participates in the regulation of inflammatory gene expression; however, if PI3K is inhibited macrophages are unable to mount an inflammatory antiviral response and die by apoptosis.


Subject(s)
Cardiovirus Infections/immunology , Encephalomyocarditis virus/immunology , Macrophage Activation/immunology , Phosphatidylinositol 3-Kinases/immunology , RNA, Double-Stranded/immunology , Animals , Cardiovirus Infections/enzymology , Caspase 3/immunology , Cell Line , Cyclooxygenase 2/immunology , Encephalomyocarditis virus/enzymology , Interleukin-1beta/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/virology , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/immunology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology
17.
Virology ; 383(1): 86-92, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-18977501

ABSTRACT

Suitable animal models are needed to study monkeypox virus (MPXV) as human monkeypox clinically resembles smallpox and MPXV is a zoonotic and potential bioterroristic agent. We have demonstrated that a species of African dormice, Graphiurus kelleni, is susceptible to a lethal infection of MPXV and that MPXV replicated in multiple organs of this species. Following intranasal administration, MPXV replicated locally in the nasal mucosa causing necrosis and hemorrhage with subsequent systemic spread to lymph nodes, spleen, liver, and other tissues where it caused severe necrosis and/or hemorrhage leading to death. The dormouse model was validated for testing prophylactic (Dryvax vaccine) and therapeutic (cidofovir) test articles against intranasal challenges with MPXV.


Subject(s)
Disease Models, Animal , Monkeypox virus/growth & development , Mpox (monkeypox)/virology , Myoxidae/virology , Animal Structures/pathology , Animal Structures/virology , Animals , Antiviral Agents/therapeutic use , Cidofovir , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Hemorrhage , Humans , Mpox (monkeypox)/drug therapy , Mpox (monkeypox)/immunology , Mpox (monkeypox)/pathology , Necrosis , Organophosphonates/therapeutic use , Smallpox Vaccine/immunology
18.
PLoS Pathog ; 4(12): e1000249, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19112490

ABSTRACT

Poxviruses subvert the host immune response by producing immunomodulatory proteins, including a complement regulatory protein. Ectromelia virus provides a mouse model for smallpox where the virus and the host's immune response have co-evolved. Using this model, our study investigated the role of the complement system during a poxvirus infection. By multiple inoculation routes, ectromelia virus caused increased mortality by 7 to 10 days post-infection in C57BL/6 mice that lack C3, the central component of the complement cascade. In C3(-/-) mice, ectromelia virus disseminated earlier to target organs and generated higher peak titers compared to the congenic controls. Also, increased hepatic inflammation and necrosis correlated with these higher tissue titers and likely contributed to the morbidity in the C3(-/-) mice. In vitro, the complement system in naïve C57BL/6 mouse sera neutralized ectromelia virus, primarily through the recognition of the virion by natural antibody and activation of the classical and alternative pathways. Sera deficient in classical or alternative pathway components or antibody had reduced ability to neutralize viral particles, which likely contributed to increased viral dissemination and disease severity in vivo. The increased mortality of C4(-/-) or Factor B(-/-) mice also indicates that these two pathways of complement activation are required for survival. In summary, the complement system acts in the first few minutes, hours, and days to control this poxviral infection until the adaptive immune response can react, and loss of this system results in lethal infection.


Subject(s)
Complement System Proteins/physiology , Ectromelia, Infectious/immunology , Ectromelia, Infectious/mortality , Animals , Complement C3/genetics , Complement C4/genetics , Complement Factor B/genetics , Complement System Proteins/genetics , Ectromelia, Infectious/genetics , Ectromelia, Infectious/pathology , Hepatitis, Viral, Animal/genetics , Hepatitis, Viral, Animal/pathology , Hepatitis, Viral, Animal/virology , Immunity, Innate/genetics , Liver/pathology , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Necrosis/genetics , Necrosis/pathology , Necrosis/veterinary , Necrosis/virology , Viremia/mortality , Virus Replication/genetics
19.
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
20.
J Gen Virol ; 89(Pt 3): 751-759, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18272767

ABSTRACT

Natural killer (NK) cells are known for their ability to lyse tumour cell targets. Studies of infections by a number of viruses, including poxviruses and herpesviruses, have demonstrated that NK cells are vital for recovery from these infections. Little is known of the ability of viruses to infect and complete a productive replication cycle within NK cells. Even less is known concerning the effect of infection on NK cell biology. This study investigated the ability of ectromelia virus (ECTV) to infect NK cells in vitro and in vivo. Following ECTV infection, NK cell gamma interferon (IFN-gamma) production was diminished and infected cells ceased proliferating and lost viability. ECTV infection of NK cells led to early and late virus gene expression and visualization of immature and mature virus particles, but no detectable increase in viable progeny virus. It was not unexpected that early gene expression occurred in infected NK cells, as the complete early transcription system is packaged within the virions. The detection of the secreted early virus-encoded immunomodulatory proteins IFN-gamma-binding protein and ectromelia inhibitor of complement enzymes (EMICE) in NK cell culture supernatants suggests that even semi-permissive infection may permit immunomodulation of the local environment.


Subject(s)
Ectromelia virus/pathogenicity , Interleukin-2/immunology , Killer Cells, Natural , Lymphocyte Activation/immunology , Virus Replication , Animals , Cells, Cultured , Ectromelia virus/physiology , Ectromelia, Infectious/virology , Female , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...