Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Virus Res ; 244: 164-172, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29113825

ABSTRACT

In 2007, numerous hamadryas baboons (Papio hamadryas) died suddenly in an aviary of a primate institute in Sochi, Russia, in the absence of prior clinical signs. Necropsies were suggestive of encephalomyocarditis virus infection, but RT-PCR assays with commonly used primers were negative. Here we report the histopathological results obtained during necropsies and the isolation and genomic characterization of a divergent strain of encephalomyocarditis virus 1 (EMCV-1) from heart tissue of one of the succumbed hamadryas baboons. Phylogenetic analysis indicates that the isolated virus belongs to the newly proposed EMCV-1 lineage G, which clusters alongside lineage C ("Mengo virus"). This study is the first report describing a lineage G strain of EMCV-1 as the etiological agent of a lethal disease outbreak among captive nonhuman primates in Europe.


Subject(s)
Cardiovirus Infections/epidemiology , Disease Outbreaks , Encephalomyocarditis virus/genetics , Genome, Viral , Papio hamadryas/virology , RNA, Viral/genetics , Amino Acid Sequence , Animals , Animals, Zoo , Autopsy , Cardiovirus Infections/mortality , Cardiovirus Infections/pathology , Cardiovirus Infections/virology , DNA, Complementary/chemistry , DNA, Complementary/genetics , Encephalomyocarditis virus/classification , Encephalomyocarditis virus/isolation & purification , Encephalomyocarditis virus/pathogenicity , Heart/virology , Phylogeny , Russia/epidemiology , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid
2.
Immunobiology ; 222(2): 350-357, 2017 02.
Article in English | MEDLINE | ID: mdl-27665995

ABSTRACT

Viral infections can give rise to secondary bacterial infections. In the present study, we examined the role of invariant natural killer T (iNKT) cells in lipopolysaccharide (LPS)-induced lethal shock during encephalomyocarditis virus (EMCV) infection. Wild-type (WT) mice and Jα18 gene knockout (Jα18 KO) mice were inoculated with EMCV, 5days prior to challenging with LPS. The survival rate of Jα18 KO mice subjected to EMCV and LPS was significantly higher than that of WT mice. TNF-α and nitric oxide (NO) production were increased in WT mice, than that in Jα18 KO mice, after the administration of EMCV and LPS. EMCV infection increased the number of iNKT cells and IFN-γ production by iNKT cells in WT mice. Moreover, EMCV infection enhanced the expression of Toll-like receptor 4 (TLR4) in the lung and spleen. IFN-γ also increased the expression of TLR4 in splenocytes. These findings indicated that EMCV infection activated iNKT cells, and IFN-γ secreted from the iNKT cells up-regulated the expression of TLR4 in various tissues. As a result, EMCV-infected mice were susceptible to LPS and easily developed the lethal shock. In conclusion, iNKT cells were involved in the development of LPS-induced lethal shock during EMCV infection.


Subject(s)
Cardiovirus Infections/immunology , Cardiovirus Infections/metabolism , Encephalomyocarditis virus/immunology , Lipopolysaccharides/adverse effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Shock, Septic/etiology , Shock, Septic/metabolism , Animals , Biomarkers , Cardiovirus Infections/mortality , Cardiovirus Infections/virology , Coinfection , Cytokines/metabolism , Disease Models, Animal , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Nitric Oxide/metabolism , Shock, Septic/mortality , Tumor Necrosis Factor-alpha/metabolism
3.
Infect Genet Evol ; 30: 19-26, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25497352

ABSTRACT

Encephalomyocarditis virus (EMCV) is a small non-enveloped, single-stranded RNA virus. It can infect many host species and cause acute myocarditis and respiratory failure in piglets, reproductive failure in pregnant sows. Diseases caused by EMCV currently affect the swine industry worldwide. In this study, an EMCV strain was isolated from an aborted fetus in western China. It was identified by reverse-transcription polymerase chain reaction (RT-PCR) and genome sequencing. The subsequent results showed that the virus could produce a specific cytopathic effect on BHK-21 cells and could cause severe clinical symptoms and pathological changes in mice. Complete genome sequencing and multiple sequence alignment indicated that the GS01 strain was 79.9-99.9% identical with other isolates worldwide. Phylogenetic analysis showed that EMCV isolates fell into five clusters: lineage 1, 2, 3, 4, and 5 based on the nucleotide sequences of the entire ORF and VP3/VP1 junction, as well as 3D gene. GS01 isolate was grouped into lineage 1. The results of this study confirmed that an EMCV strain GS01 isolated from an aborted pig fetus in western China was fatal to mice and provided new epidemiologic data on EMCV in China.


Subject(s)
Cardiovirus Infections/virology , Encephalomyocarditis virus/classification , Encephalomyocarditis virus/genetics , Swine Diseases/virology , Animals , Cardiovirus Infections/mortality , Cardiovirus Infections/veterinary , Cell Line , China , Cricetinae , Encephalomyocarditis virus/isolation & purification , Mice , Phylogeny , Swine/virology
4.
J Neuroimmunol ; 277(1-2): 118-26, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25468274

ABSTRACT

Viruses use various mechanisms to evade clearance by the host. Investigating how a few changes in the genome of a non-lethal virus can lead to altered disease, from survivable to immunosuppression/death, would provide valuable information into viral pathogenesis. The Daniels strain of Theiler's murine encephalomyelitis virus causes an asymptomatic infection or acute encephalitis followed by viral clearance. A mutant, H101, carries several alterations in the viral genome. H101 infection causes profound immunosuppression and death. Thus, a virus that is normally cleared by its natural host can become lethal due to just a few changes in the viral genome.


Subject(s)
Cardiovirus Infections/pathology , Immunosuppression Therapy/adverse effects , Mutation/genetics , Theilovirus/genetics , Theilovirus/pathogenicity , Animals , Antigens, CD/metabolism , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Female , Freund's Adjuvant/toxicity , Male , Mice , Mice, Inbred C57BL , Myelin Proteolipid Protein/immunology , Myelin Proteolipid Protein/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity , Phycocyanin/metabolism , Spleen/pathology , Time Factors
5.
Mol Ther ; 21(5): 1087-95, 2013 May.
Article in English | MEDLINE | ID: mdl-23568262

ABSTRACT

Picornaviruses have been developed as potential therapies for gene delivery and vaccination. One drawback to their use is the potential for recombination and viral persistence. Therefore, the engineering strategies used must take into account the possibility for virus escape. We have developed Theiler's murine encephalomyelitis virus (TMEV) as a potential vaccine vector for use in immunotherapy. This study shows that insertion of a vaccine epitope at a unique site within the TMEV leader protein can dramatically increase the type I interferon (IFN) response to infection and promote rapid viral clearance. This live virus vaccine maintains its ability to drive antigen-specific CD8(+) T-cell responses to a model antigen as well as to the weakly immunogenic tumor antigen Her2/neu. Furthermore, the epitope integration site does not affect the efficacy of this vaccine as cancer immunotherapy for treating models of melanoma and breast cancer as demonstrated by delayed tumor outgrowth and increased survival in animals implanted with these tumors. These findings show that an attenuated virus retaining limited ability to replicate nonetheless can effectively mobilize CD8(+) cellular immunity and will be important for the design of picornavirus vectors used as immunotherapy in clinical settings.


Subject(s)
Antigens/immunology , Cancer Vaccines/immunology , Epitopes/immunology , Neoplasms/immunology , Theilovirus/immunology , Amino Acid Sequence , Animals , Base Sequence , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Cardiovirus Infections/virology , Cell Line, Tumor , Female , Genetic Vectors/genetics , Genetic Vectors/immunology , Humans , Immunotherapy , Interferon Type I/immunology , Mice , Molecular Sequence Data , Mutagenesis, Insertional , Neoplasms/pathology , Neoplasms/therapy , Receptor, ErbB-2/immunology , Theilovirus/genetics , Tumor Burden/drug effects , Vaccines, Attenuated , Viral Proteins/chemistry , Viral Proteins/immunology
6.
Nat Immunol ; 13(11): 1055-62, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23042151

ABSTRACT

Toll-like receptors (TLRs) sense pathogen-associated molecules and respond by inducing cytokines and type I interferon. Here we show that genetic ablation of the E3 ubiquitin ligase Pellino3 augmented the expression of type I interferon but not of proinflammatory cytokines in response to TLR3 activation. Pellino3-deficient mice had greater resistance against the pathogenic and lethal effects of encephalomyocarditis virus (EMCV). TLR3 signaling induced Pellino3, which in turn interacted with and ubiquitinated TRAF6. This modification suppressed the ability of TRAF6 to interact with and activate IRF7, resulting in downregulation of type I interferon expression. Our findings highlight a new physiological role for Pellino3 and define a new autoregulatory network for controlling type I interferon expression.


Subject(s)
Cardiovirus Infections/immunology , Gene Expression Regulation , Interferon Type I/immunology , Toll-Like Receptor 3/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/mortality , Cardiovirus Infections/virology , Encephalomyocarditis virus/immunology , Homeostasis , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Interferon Type I/genetics , Mice , Mice, Knockout , Signal Transduction , Survival Rate , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , Toll-Like Receptor 3/genetics , Ubiquitin/genetics , Ubiquitin/immunology , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
7.
Sci Rep ; 2: 367, 2012.
Article in English | MEDLINE | ID: mdl-22509465

ABSTRACT

Secondary bacterial infection in humans is one of the pathological conditions requiring clinical attention. In this study, we examined the effect of lipopolysaccharide (LPS) on encephalomyocarditis virus (EMCV) infected mice. All mice inoculated with EMCV at 5 days before LPS challenge died within 24 h. LPS-induced TNF-α mRNA expression was significantly increased in the brain and heart at 5 days after EMCV infection. CD11b(+)/TLR4(+) cell population in the heart was remarkably elevated at 5 days after EMCV infection, and sorted CD11b(+) cells at 5 days after EMCV infection produced a large amount of TNF-α on LPS stimulation in vivo and in vitro. In conclusion, we found that the infiltration of CD11b(+) cells into infected organs is involved in the subsequent LPS-induced lethal shock in viral encephalomyocarditis. This new experimental model can help define the mechanism by which secondary bacterial infection causes a lethal shock in viral encephalomyocarditis.


Subject(s)
Cardiovirus Infections/complications , Encephalomyocarditis virus/pathogenicity , Lipopolysaccharides/toxicity , Animals , Base Sequence , CD11b Antigen/immunology , Cardiovirus Infections/immunology , Cardiovirus Infections/metabolism , Cardiovirus Infections/mortality , DNA Primers , Flow Cytometry , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Immunol ; 188(8): 3980-7, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22422885

ABSTRACT

The activity of IDO that catalyzes the degradation of tryptophan (Trp) into kynurenine (Kyn) increases after diseases caused by different infectious agents. Previously, we demonstrated that IDO has an important immunomodulatory function in immune-related diseases. However, the pathophysiological role of IDO following acute viral infection is not fully understood. To investigate the role of IDO in the l-Trp-Kyn pathway during acute viral myocarditis, mice were infected with encephalomyocarditis virus, which induces acute myocarditis. We used IDO-deficient (IDO(-/-)) mice and mice treated with 1-methyl-d,l-Trp (1-MT), an inhibitor of IDO, to study the importance of Trp-Kyn pathway metabolites. Postinfection with encephalomyocarditis virus infection, the serum levels of Kyn increased, whereas those of Trp decreased, and IDO activity increased in the spleen and heart. The survival rate of IDO(-/-) or 1-MT-treated mice was significantly greater than that of IDO(+/+) mice. Indeed, the viral load was suppressed in the IDO(-/-) or 1-MT-treated mice. Furthermore, the levels of type I IFNs in IDO(-/-) mice and IDO(-/-) bone marrow-transplanted IDO(+/+) mice were significantly higher than those in IDO(+/+) mice, and treatment of IDO(-/-) mice with Kyn metabolites eliminated the effects of IDO(-/-) on the improved survival rates. These results suggest that IDO has an important role in acute viral myocarditis. Specifically, IDO increases the accumulation of Kyn pathway metabolites, which suppress type I IFNs production and enhance viral replication. We concluded that inhibition of the Trp-Kyn pathway ameliorates acute viral myocarditis.


Subject(s)
Cardiovirus Infections/immunology , Encephalomyocarditis virus , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Kynurenine/immunology , Myocarditis/immunology , Myocardium/immunology , Tryptophan/immunology , Acute Disease , Animals , Cardiovirus Infections/mortality , Cardiovirus Infections/virology , Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon Type I/biosynthesis , Interferon Type I/immunology , Kynurenine/blood , Male , Mice , Mice, Knockout , Myocarditis/mortality , Myocarditis/virology , Myocardium/metabolism , Signal Transduction/immunology , Spleen/immunology , Spleen/metabolism , Spleen/virology , Survival Rate , Tryptophan/analogs & derivatives , Tryptophan/blood , Tryptophan/pharmacology , Viral Load/drug effects , Virus Replication
9.
J Virol ; 85(8): 3708-16, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21307205

ABSTRACT

Induction of apoptosis in cells infected by Sendai virus (SeV), which triggers the cytosolic RIG-I pathway, requires the presence of interferon regulatory factor 3 (IRF-3). Independent of IRF-3's transcriptional role, a novel IRF-3 activation pathway causes its interaction with the proapoptotic protein Bax and its mitochondrial translocation to induce apoptosis. Here we report that two other RNA viruses, vesicular stomatitis virus (VSV) and encephalomyocarditis virus (EMCV), may also activate the same pathway. Moreover, cytosolic DNA, produced by adenovirus or introduced by transfection, activated the pathway in an RNA polymerase III-dependent fashion. To evaluate the contribution of this newly discovered apoptotic pathway to the host's overall antiviral response, we measured the efficiencies of replication of various viruses in vitro and viral pathogenesis in vivo, using cells and mice that are selectively deficient in components required for the apoptotic pathway of IRF-3. Our results clearly demonstrate that the IRF-3/Bax-mediated apoptotic signaling branch contributes significantly to the host's protection from viral infection and consequent pathogenesis.


Subject(s)
Apoptosis , DNA, Viral/metabolism , Interferon Regulatory Factor-3/immunology , RNA, Viral/metabolism , Virus Replication , bcl-2-Associated X Protein/immunology , Adenoviridae/immunology , Animals , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Cell Line , Disease Models, Animal , Encephalomyocarditis virus/immunology , Interferon Regulatory Factor-3/deficiency , Mice , Mice, Knockout , Sendai virus/immunology , Survival Analysis , Vesicular Stomatitis/immunology , bcl-2-Associated X Protein/deficiency
10.
J Virol ; 85(1): 621-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20962089

ABSTRACT

Transgenic expression of the RNA-dependent RNA polymerase 3D(pol) inhibited infection of Theiler's murine encephalitis virus (TMEV), a picornavirus from which it was derived. Here, we infected 3D(pol) transgenic mice with another picornavirus, as well as an alphaherpesvirus and a rhabdovirus. 3D(pol) transgenic FVB mice had significantly lower viral loads and survived longer after infection with all three types of viruses than nontransgenic FVB mice. Viral inhibition among three different types of virus by transgenic 3D(pol) suggests that the mechanism of action is not the direct interference with picornaviral 3D(pol) but instead may be the changing of host cells to an antiviral state before or after viral infection occurs, as basal interferon levels were higher in 3D(pol) transgenic mice before infection. Further study of this mechanism may open new possibilities for future antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Herpesvirus 1, Suid/drug effects , RNA-Dependent RNA Polymerase/pharmacology , Theilovirus/enzymology , Vesicular stomatitis Indiana virus/drug effects , Animals , Antiviral Agents/metabolism , Brain/pathology , Brain/virology , Cardiovirus Infections/mortality , Cardiovirus Infections/virology , Encephalomyocarditis virus/pathogenicity , Herpesvirus 1, Suid/pathogenicity , Mice , Mice, Transgenic , Pseudorabies/mortality , Pseudorabies/virology , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Rhabdoviridae Infections/mortality , Rhabdoviridae Infections/virology , Theilovirus/genetics , Vesicular stomatitis Indiana virus/pathogenicity
11.
Physiol Behav ; 95(1-2): 63-71, 2008 Sep 03.
Article in English | MEDLINE | ID: mdl-18538803

ABSTRACT

Previous research has shown that chronic restraint stress exacerbates Theiler's virus infection, a murine model for CNS inflammation and multiple sclerosis. The current set of experiments was designed to evaluate the potential role of glucocorticoids in the deleterious effects of restraint stress on acute CNS inflammatory disease. Exposure to chronic restraint stress resulted in elevated levels of corticosterone as well as increased clinical scores and weight loss (Experiment 1). In addition, corticosterone administration alone exacerbated behavioral signs of TMEV-induced sickness (i.e. decreased body weight, increased symptoms of encephalitis, and increased mortality) and reduced inflammation in the CNS (Experiment 2). Infected subjects receiving exogenous corticosterone showed exacerbation of acute phase measures of sickness and severe mortality as well as decreased viral clearance from CNS (Experiment 3). These findings indicate that corticosterone exposure alone is sufficient to exacerbate acute CNS inflammatory disease.


Subject(s)
Cardiovirus Infections/etiology , Cardiovirus Infections/physiopathology , Glucocorticoids/administration & dosage , Theilovirus/pathogenicity , Animals , Body Weight/drug effects , Body Weight/physiology , Cardiovirus Infections/metabolism , Cardiovirus Infections/mortality , Central Nervous System/drug effects , Central Nervous System/pathology , Central Nervous System/virology , Glucocorticoids/metabolism , Male , Meningitis/etiology , Meningitis/pathology , Meningitis/virology , Mice , Mice, Inbred CBA , Mortality , Severity of Illness Index , Stress, Psychological/physiopathology , Survival Analysis
12.
Indian J Exp Biol ; 43(12): 1156-60, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16359127

ABSTRACT

The ethanolic extracts, various fractions and two pure compounds isolated from the plant N. arbortris were tested against Encephalomyocarditis Virus (EMCV) and Semliki Forest Virus (SFV). Pronounced in vitro virus inhibitory activity was observed with the ethanolic and n-butanol fractions as well as with the pure compounds arbortristoside A and arbortristoside C. In addition, ethanolic extracts and n-butanol fraction protected EMCV infected mice to the extent of 40 and 60% respectively against SFV at a daily dose of 125 mg/kg body weight.


Subject(s)
Encephalomyocarditis virus/drug effects , Oleaceae , Seeds , Semliki forest virus/drug effects , 1-Butanol/administration & dosage , 1-Butanol/pharmacology , Administration, Oral , Alphavirus Infections/drug therapy , Alphavirus Infections/mortality , Animals , Cardiovirus Infections/drug therapy , Cardiovirus Infections/mortality , Chlorocebus aethiops , Dose-Response Relationship, Drug , Glycosides/administration & dosage , Glycosides/pharmacology , Injections, Intraperitoneal , Iridoids/administration & dosage , Iridoids/pharmacology , Mice , Phytotherapy/methods , Plant Extracts/pharmacology , Vero Cells
13.
Am J Physiol Heart Circ Physiol ; 289(4): H1577-83, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15923319

ABSTRACT

Recent studies point to important interactions between proinflammatory cytokines and neurohumoral mediators in heart failure. Here we investigate the influence of the beta-adrenergic system on cytokines and neurohumoral factors and the sequelae of viral myocarditis. In an experimental model with virus-infected BALB/c mice, we studied the acute and chronic effects of epinephrine and propranolol on myocardial morphology, cytokine gene expression, and survival. BALB/c mice were inoculated with the encephalomyocarditis virus (EMCV) or sham inoculated with saline and followed for 30 days. Epinephrine increased the severity of inflammatory cell infiltration and myocardial necrosis induced by EMCV. Gene expression of TNF-alpha, IL-6, and IL-10 was markedly enhanced by epinephrine in EMCV-inoculated mice. Survival rate after 30 days was reduced to 40% in epinephrine-treated EMCV-inoculated mice compared with 70% in untreated EMCV-inoculated mice (P < 0.05). Treatment with the beta-blocker propranolol significantly decreased mortality, myocardial necrosis, and infiltration of inflammatory cells in EMCV-inoculated mice. Propranolol also suppressed gene expression of TNF-alpha, IL-6, and IL-10. A single dose of epinephrine 120 days after EMCV inoculation caused sudden death in 70% of infected mice; propranolol significantly reduced incidence of death to 33%. These results indicate that acute and chronic stages of viral myocarditis are modulated by the beta-adrenergic system and its interactions with proinflammatory cytokines.


Subject(s)
Adrenergic Agonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cardiovirus Infections/drug therapy , Encephalomyocarditis virus , Epinephrine/pharmacology , Myocarditis/drug therapy , Propranolol/pharmacology , Acute Disease , Amnion/cytology , Animals , Arrhythmias, Cardiac/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Chronic Disease , Cytokines/genetics , Death, Sudden, Cardiac/prevention & control , Gene Expression/immunology , Humans , Male , Mice , Mice, Inbred BALB C , Myocarditis/mortality , Myocarditis/virology
14.
J Immunol ; 174(11): 6854-62, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905527

ABSTRACT

Peptide immunotherapy both activates and suppresses the T cell response against known peptide Ags. Although pretreatment with VP2(121-130) peptide inhibits the development of antiviral CTL specific for the immunodominant D(b):VP2(121-130) epitope expressed during acute Theiler's murine encephalomyelitis virus infection, i.v. injection of this same peptide or MHC tetramers containing the peptide during an ongoing antiviral CTL response results in a peptide-induced fatal syndrome (PIFS) within 48 h. Susceptibility to PIFS is dependent on peptide-specific CD8(+) T cells, varies among inbred strains of mice, and is not mediated by traditionally defined mechanisms of shock. Analyses using bone marrow chimeras and mutant mice demonstrate that susceptibility to PIFS is determined by the genotype of bone marrow-derived cells and requires the expression of perforin. Animals responding to peptide treatment with PIFS develop classical stress responses in the brain. These findings raise important considerations for the development of peptide therapies for active diseases to modify immune responses involving expanded populations of T cells. In summary, treatment with peptides or MHC-tetramers during a peptide-specific immune response can result in a fatal shock-like syndrome. Susceptibility to the syndrome is genetically determined, is mediated by CD8(+) T cells, and requires expression of perforin. These findings raise concerns about the use of peptides and MHC tetramers in therapeutic schemes.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Capsid Proteins/adverse effects , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Epitopes, T-Lymphocyte/adverse effects , Immunodominant Epitopes/adverse effects , Theilovirus/immunology , Animals , Antigen Presentation/genetics , CD8-Positive T-Lymphocytes/metabolism , Capillary Permeability/genetics , Capillary Permeability/immunology , Capsid Proteins/administration & dosage , Capsid Proteins/immunology , Cardiovirus Infections/genetics , Cardiovirus Infections/physiopathology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Female , Genetic Predisposition to Disease/genetics , H-2 Antigens/immunology , Histocompatibility Antigen H-2D , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/physiology , Immunity, Innate/genetics , Immunization Schedule , Immunodominant Epitopes/administration & dosage , Immunodominant Epitopes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Peptide Fragments/administration & dosage , Peptide Fragments/adverse effects , Peptide Fragments/immunology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Syndrome , Interferon gamma Receptor
15.
J Virol ; 76(5): 2217-24, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11836399

ABSTRACT

During the first 45 days after intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV), the levels of mRNAs encoding chemokines MCP-1/CCL2, RANTES/CCL5, and IP-10/CXCL10 in the central nervous system (CNS) are closely related to the sites of virus gene expression and tissue inflammation. In the present study, these chemokines were monitored during the latter 135 days of a 6-month course of TMEV-induced disease in susceptible (PLJ) or resistant (C57BL/6) mice that possessed or lacked either CD4+ or CD8+ T cells. These data were additionally correlated to mouse genotype, virus persistence in the CNS, antiviral antibody titers, mortality, and the severity of neurological disease. Surprisingly, the major determinant of chemokine expression was virus persistence: the factors of susceptible or resistant genotype, severity of neuropathology, and presence or absence of regulatory T cells exerted minimal effects. Our observations indicated that chemokine expression in the CNS in this chronic viral disorder was intrinsic to the CNS innate immune response to infection and was not governed by elements of the adaptive immune system.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/metabolism , Chemokines/metabolism , Multiple Sclerosis/virology , Theilovirus/physiology , Animals , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Cardiovirus Infections/physiopathology , Cardiovirus Infections/virology , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/immunology , Multiple Sclerosis/mortality , Multiple Sclerosis/physiopathology , Spinal Cord/metabolism , Theilovirus/immunology , Theilovirus/pathogenicity , Virus Replication
16.
Cell Transplant ; 11(8): 753-8, 2002.
Article in English | MEDLINE | ID: mdl-12588107

ABSTRACT

We used mice to test our hypothesis that in response to viral invasion, stem cells may migrate into the heart and attenuate the effect of viral myocarditis. Male BALB/c mice were divided into three groups: mouse embryonic stem (ES) cell control, encephalomyocarditis virus (EMCV), and EMCV + ES cells. After administration of ES cells via tail vein, mice were immediately inoculated with EMCV. Mice were sacrificed at different days after EMCV inoculation. Mortality was recorded. Inflammatory cell infiltration and necrosis (major pathological changes of viral myocarditis) were evaluated by hematoxylin-eosin staining. ES cell migration and differentiation were identified by immunofluorescence. The survival rate in the EMCV + ES cell group (80%) was significantly increased (p < 0.05) over the EMCV-alone group (64%). Also, the incidence of inflammatory cell infiltration and myocardial lesions was lower in the EMCV + ES cell mice. Furthermore, the result of green fluorescent protein (GFP) and alpha-actinin analysis indicated that ES cells migrated into the heart and differentiated into myocytes after virus inoculation. In conclusion, ES cells significantly increased the survival of viral myocarditis mice and also decreased the necrosis and infiltration of inflammatory cells. These results demonstrated the ability of stem cells to mitigate the effects of viral infection on the heart and illustrated their potential therapeutic application to other mammalian species, including humans.


Subject(s)
Cardiovirus Infections/therapy , Encephalomyocarditis virus , Myocarditis/therapy , Stem Cell Transplantation , Animals , Cardiovirus Infections/mortality , Cell Differentiation , Cell Line , Cell Movement , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Myocarditis/mortality , Myocarditis/virology , Myocytes, Cardiac/cytology , Stem Cells/cytology
17.
Brain Behav Immun ; 15(3): 235-54, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566048

ABSTRACT

Restraint stress was found to have a profound effect on the acute phase of Theiler's virus infection. Increased mortality rates were observed in restrained CBA mice infected with the BeAn strain of Theiler's virus. In addition, restrained mice developed higher CNS viral titers than infected/nonrestrained mice. Thymic atrophy was observed in both infected and uninfected restrained mice. Decreased microgliosis, perivascular cuffing, and astrocytosis were observed in restrained mice compared to nonrestrained infected mice at 7 days postinfection. Restraint-stressed mice also developed decreased numbers of lymphocytes and increased numbers of neutrophils in the blood. The mechanism proposed for these alterations involves stress-induced corticosterone, which causes immunosuppression, decreased trafficking of inflammatory cells in the CNS, and, consequently, increased viral replication.


Subject(s)
Cardiovirus Infections/complications , Cardiovirus Infections/etiology , Central Nervous System Diseases/virology , Restraint, Physical , Theilovirus , Acute Disease , Animals , Cardiovirus Infections/mortality , Cardiovirus Infections/physiopathology , Central Nervous System/pathology , Central Nervous System Diseases/mortality , Central Nervous System Diseases/pathology , Central Nervous System Diseases/physiopathology , Corticosterone/blood , Leukocyte Count , Male , Mice , Mice, Inbred CBA , Thymus Gland/pathology
18.
J Interferon Cytokine Res ; 21(8): 575-81, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11559435

ABSTRACT

Intraperitoneal (i.p.) administration of 20,000 IU recombinant murine IFN-alpha (rMuIFN-alpha) was highly effective in protecting mice challenged i.p. with doses of encephalomyocarditis virus (EMCV) ranging from 44 to 440 LD(50) (p<0.001). Oromucosal (o.m.) IFN therapy was also found to be effective in protecting mice challenged with a lethal dose of EMCV. Thus, 40% of animals infected with 44 LD(50) of EMCV and treated o.m. with 20,000 IU rMuIFN-alpha survived infection with a mean survival time of 12.0 +/- 2.46 days relative to a mean of 6.11 +/- 0.38 days in the control group (p<0.05). Oromucosal IFN therapy was found to be ineffective, however, in animals infected with higher doses of EMCV (88-440 LD(50)), even though intraperitoneal administration of the same dose of rMuIFN-alpha resulted in the survival of 90%, 50%, and 60% of animals infected with 88, 220, and 440 LD(50) of EMCV, respectively. These results suggest that oromucosal IFN therapy is effective at relatively low viral load only and that the mechanism of action of oromucosal IFN therapy may be different from that of parenterally administered IFN. Our results suggest that oromucosal IFN therapy may be most effective in chronic viral infections as an alternative to parenterally administered IFN, which is clinically effective but poorly tolerated.


Subject(s)
Antiviral Agents/administration & dosage , Cardiovirus Infections/drug therapy , Cardiovirus Infections/virology , Interferon Type I/administration & dosage , Viral Load , Administration, Intranasal , Animals , Antiviral Agents/therapeutic use , Cardiovirus Infections/mortality , Encephalomyocarditis virus/drug effects , Injections, Intraperitoneal , Interferon Type I/therapeutic use , Lethal Dose 50 , Male , Mice , Mouth Mucosa/drug effects , Mouth Mucosa/virology , Oropharynx , Recombinant Proteins , Survival Rate
19.
J Interferon Cytokine Res ; 21(7): 539-45, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11506749

ABSTRACT

Interferons (IFN) have been shown to be effective in protecting animals against lethal viral infections when administered systemically in relatively high doses. Intraperitoneal (i.p.) injection of mice with encephalomyocarditis virus (EMCV) gives rise to a rapidly progressive fatal disease characterized by central nervous system involvement and encephalitis. IFN-alpha has been shown to be effective in protecting mice against lethal EMCV infection when given via parenteral and oral/sublingual routes. The current study was designed to explore the ability of orally/sublingually and intranasally (i.n.) administered IFN-alpha to treat mice infected with EMCV in support of a planned clinical trial to evaluate efficacy of oral IFN-alpha in human viral infections. The primary objective of the study was to determine the efficacy of recombinant murine IFN-alpha (rMuIFN-alpha) in the treatment of mice infected with 100 LD(50) EMCV following oral, i.n., and i.p. administration at doses of 20,000 and 100,000 IU. The results of the current experiment did not indicate protection from infection with EMCV in mice that received IFN by the i.n. or oral/sublingual routes. The negative controls, infection of mice with 100 LD(50) of EMCV followed by treatment with excipient via all three routes, resulted in death of nearly all mice, as expected. The positive control, treatment of EMCV-infected (100 LD(50)) mice with rMuIFN-alpha via the i.p. route, was successful in protecting a significant number of mice from death compared with matched controls. This study points out the need to determine the optimum conditions for administration of oral/sublingual or i.n. IFN to insure maximum efficacy against viral infections.


Subject(s)
Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Cardiovirus Infections/drug therapy , Encephalomyocarditis virus/drug effects , Interferon Type I/administration & dosage , Interferon Type I/adverse effects , Administration, Intranasal , Administration, Oral , Animals , Antiviral Agents/therapeutic use , Cardiovirus Infections/mortality , Drug Evaluation, Preclinical/methods , Female , Injections, Intraperitoneal , Interferon Type I/therapeutic use , Lethal Dose 50 , Mice , Random Allocation , Recombinant Proteins , Titrimetry
20.
Mol Cell Neurosci ; 15(6): 495-509, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10860577

ABSTRACT

The importance of genetic susceptibility in determining the progression of demyelination and neurologic deficits is a major focus in neuroscience. We studied the influence of human leukocyte antigen (HLA)-DQ polymorphisms on disease course and neurologic impairment in virus-induced demyelination. HLA-DQ6 or DQ8 was inserted as a transgene into mice lacking endogenous expression of MHC class I (beta(2)m) and class II (H2-A(beta)) molecules. Following Theiler's murine encephalomyelitis virus (TMEV) infection, we assessed survival, virus persistence, demyelination, and clinical disease. Mice lacking expression of endogenous class I and class II molecules (beta(2)m(o) Abeta(o) mice) died 3 to 4 weeks postinfection (p.i.) due to overwhelming virus replication in neurons. beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice had increased survival and decreased gray matter disease and virus replication compared to nontransgenic littermate controls. Both beta(2)m(o) Abeta(o) DQ6 and beta(2)m(o) Abeta(o) DQ8 mice developed chronic virus persistence in glial cells of the white matter of the spinal cord, with greater numbers of virus antigen-positive cells in beta(2)m(o) Abeta(o) DQ8 than in beta(2)m(o) Abeta(o) DQ6 mice. At day 45 p.i., the demyelinating lesions in the spinal cord of beta(2)m(o) Abeta(o) DQ8 were larger than those in the beta(2)m(o) Abeta(o) DQ6 mice. Earlier and more profound neurologic deficits were observed in beta(2)m(o) Abeta (o) DQ8 mice compared to beta(2)m(o) Abeta(o) DQ6 mice, although by 120 days p.i. both strains of mice showed similar extent of demyelination and neurologic deficits. Delayed-type hypersensitivity and antibody responses to TMEV demonstrated that the mice mounted class II-mediated cellular and humoral immune responses. The results are consistent with the hypothesis that rates of progression of demyelination and neurologic deficits are related to the differential ability of DQ6 and DQ8 transgenes to modulate the immune response and control virus.


Subject(s)
Cardiovirus Infections/genetics , HLA-DQ Antigens/genetics , Multiple Sclerosis/genetics , Polymorphism, Genetic , Theilovirus , Acute Disease , Animals , Antibody Formation/genetics , Antigens, Viral/analysis , Brain/immunology , Brain/virology , Cardiovirus Infections/immunology , Cardiovirus Infections/mortality , Chronic Disease , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Disease Models, Animal , Disease Progression , Genetic Predisposition to Disease , Humans , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity , Multiple Sclerosis/immunology , Nerve Fibers/immunology , Nerve Fibers/virology , Postural Balance , Spinal Cord/immunology , Spinal Cord/virology , Survival Analysis , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...