Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Virology ; 429(2): 163-70, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22579352

ABSTRACT

DNAM-1 gene-deficient (-/-) mice take significantly longer to clear an acute and persistent LCMV infection in vivo than DNAM-1 +/+ mice. During acute LCMV priming, at the single cell level, DNAM-1 -/- mice made significantly less cytoplasmic CD8 TNF-α and IL-2 but not IFN-γ than their DNAM-1 +/+ counterparts. Restimulated immune memory CD8 T cells from DNAM-1 -/- and DNAM-1 +/+ mice were equivalent in cytolytic activity against LCMV-infected target cells but DNAM-1 -/- CD8 T cells had significant reductions in TNF-α and IL-2 that were associated on adoptive transfer with the inability to terminate the persistent viral infection.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/genetics , Arenaviridae Infections/immunology , CD8-Positive T-Lymphocytes/immunology , Interleukin-2/immunology , Lymphocytic choriomeningitis virus/immunology , Tumor Necrosis Factor-alpha/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
2.
J Virol ; 85(15): 7928-32, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21613399

ABSTRACT

The recombinant engineering of trisegmented lymphocytic choriomeningitis virus (LCMV) to express two genes of interest was recently reported. We used this technology to efficiently express green fluorescent protein (GFP) and the immunoregulatory gene product interleukin-10 (IL-10) in vitro, assess IL-10 function in vivo during viral meningitis, and generate specific, robust monoclonal antibody responses to IL-10. Tripartite viruses were attenuated in wild-type and TLR7(-/-) mice. However, IFNAR1(-/-) mice sustained systemic viral replication when 2 nucleotide substitutions from a persistent LCMV variant were present. These findings demonstrate the utility of tripartite LCMV in vitro and in vivo to study genes in the context of a well-defined model system.


Subject(s)
Antibodies, Viral/biosynthesis , Lymphocytic choriomeningitis virus/physiology , Recombination, Genetic , Viral Proteins/biosynthesis , Animals , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/metabolism , Mice , Receptor, Interferon alpha-beta/biosynthesis , Receptor, Interferon alpha-beta/genetics
3.
Proc Natl Acad Sci U S A ; 108(1): 331-6, 2011 Jan 04.
Article in English | MEDLINE | ID: mdl-21173229

ABSTRACT

Measles virus (MV), a member of the family Paramyxoviridae and an exclusively human pathogen, is among the most infectious viruses. A progressive fatal neurodegenerative complication, subacute sclerosing panencephalitis (SSPE), occurs during persistent MV infection of the CNS and is associated with biased hypermutations of the viral genome. The observed hypermutations of A-to-G are consistent with conversions catalyzed by the adenosine deaminase acting on RNA (ADAR1). To evaluate the role of ADAR1 in MV infection, we selectively disrupted expression of the IFN-inducible p150 ADAR1 isoform and found it caused embryonic lethality at embryo day (E) 11-E12. We therefore generated p150-deficient and WT mouse embryo fibroblast (MEF) cells stably expressing the MV receptor signaling lymphocyte activation molecule (SLAM or CD150). The p150(-/-) but not WT MEF cells displayed extensive syncytium formation and cytopathic effect (CPE) following infection with MV, consistent with an anti-MV role of the p150 isoform of ADAR1. MV titers were 3 to 4 log higher in p150(-/-) cells compared with WT cells at 21 h postinfection, and restoration of ADAR1 in p150(-/-) cells prevented MV cytopathology. In contrast to infection with MV, p150 disruption had no effect on vesicular stomatitis virus, reovirus, or lymphocytic choriomeningitis virus replication but protected against CPE resulting from infection with Newcastle disease virus, Sendai virus, canine distemper virus, and influenza A virus. Thus, ADAR1 is a restriction factor in the replication of paramyxoviruses and orthomyxoviruses.


Subject(s)
Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Embryonic Development/genetics , Mutation/genetics , SSPE Virus/genetics , Subacute Sclerosing Panencephalitis/genetics , Virus Replication/genetics , Animals , Antigens, CD/metabolism , Cell Line , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Knockout Techniques , Green Fluorescent Proteins , Mice , Mice, Inbred C57BL , Protein Isoforms/genetics , RNA-Binding Proteins , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signaling Lymphocytic Activation Molecule Family Member 1
4.
Virology ; 363(1): 113-23, 2007 Jun 20.
Article in English | MEDLINE | ID: mdl-17320138

ABSTRACT

Virus-specific CD4(+) T cells contribute to effective virus control through a multiplicity of mechanisms including direct effector functions as well as "help" for B cell and CD8(+) T cell responses. Here, we have used the lymphocytic choriomeningitis virus (LCMV) system to assess the minimal constraints of a dominant antiviral CD4(+) T cell response. We report that the core epitope derived from the LCMV glycoprotein (GP) is 11 amino acids in length and provides optimal recognition by epitope-specific CD4(+) T cells. Surprisingly, this epitope is also recognized by LCMV-specific CD8(+) T cells and thus constitutes a unique viral determinant with dual MHC class I- and II-restriction.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Immunodominant Epitopes/immunology , Amino Acid Sequence , Animals , Epitope Mapping , Epitopes, T-Lymphocyte/chemistry , Lymphocytic choriomeningitis virus/immunology , Mice , Molecular Sequence Data , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Cytotoxic/immunology
5.
J Proteome Res ; 5(9): 2405-16, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16944953

ABSTRACT

Mass spectrometry analysis was used to target three different aspects of the viral infection process: the expression kinetics of viral proteins, changes in the expression levels of cellular proteins, and the changes in cellular metabolites in response to viral infection. The combination of these methods represents a new, more comprehensive approach to the study of viral infection revealing the complexity of these events within the infected cell. The proteins associated with measles virus (MV) infection of human HeLa cells were measured using a label-free approach. On the other hand, the regulation of cellular and Flock House Virus (FHV) proteins in response to FHV infection of Drosophila cells was monitored using stable isotope labeling. Three complementary techniques were used to monitor changes in viral protein expression in the cell and host protein expression. A total of 1500 host proteins was identified and quantified, of which over 200 proteins were either up- or down-regulated in response to viral infection, such as the up-regulation of the Drosophila apoptotic croquemort protein, and the down-regulation of proteins that inhibited cell death. These analyses also demonstrated the up-regulation of viral proteins functioning in replication, inhibition of RNA interference, viral assembly, and RNA encapsidation. Over 1000 unique metabolites were also observed with significant changes in over 30, such as the down-regulated cellular phospholipids possibly reflecting the initial events in cell death and viral release. Overall, the cellular transformation that occurs upon viral infection is a process involving hundreds of proteins and metabolites, many of which are structurally and functionally uncharacterized.


Subject(s)
Gene Expression Regulation , Measles virus/metabolism , Nodaviridae/metabolism , Proteins/analysis , Proteomics/methods , RNA Virus Infections/metabolism , Animals , Cells, Cultured , Drosophila melanogaster , HeLa Cells , Humans , Mass Spectrometry/methods , Oxygen Isotopes
6.
Virology ; 347(1): 234-45, 2006 Mar 30.
Article in English | MEDLINE | ID: mdl-16529787

ABSTRACT

Measles virus (MV), one of the most infectious of human pathogens, still infects over 30 million humans and causes over 500,000 deaths each year [Griffin, D., 2001. Measles virus. In: Fields, B., Knipe, D., Howley, P. (Eds.), Fields Virology. Lippincott-Raven, Philadelphia, pp. 1401-1442; ]. Death is primarily due to secondary microbial infections associated with the immunosuppression caused by MV. Studies of humans with genetic or acquired deficiencies of either the humoral or cellular arm of the immune system, and rodent models have implicated T cells in the control of the ongoing MV infection but the precise role and activities of the specific T cell subset or the molecules they produce is not clear. Using a transgenic mouse model in conjunction with depletion and reconstitution of individual B and T cell subsets alone or in combination, we show that neither CD4, CD8 nor B cells per se control acute MV infection. However, combinations of either CD4 T cells and B cells, or of CD4 and CD8 T cells are essential but CD8 T with B cells are ineffective. Interferon-gamma and neutralizing antibodies, but neither perforin nor TNF-alpha alone are associated with clearance of MV infection. TNF-alpha combined with interferon-gamma is more effective in protection than interferon alone. Further, the lack of an interferon-gamma response leads to persistence of MV.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Central Nervous System Viral Diseases/immunology , Measles virus/immunology , Measles virus/pathogenicity , Measles/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System Viral Diseases/virology , Humans , Interferon-gamma/deficiency , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Cooperation , Measles/virology , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
7.
J Exp Med ; 202(9): 1185-90, 2005 Nov 07.
Article in English | MEDLINE | ID: mdl-16260490

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a progressive fatal neurodegenerative disease associated with persistent infection of the central nervous system (CNS) by measles virus (MV), biased hypermutations of the viral genome affecting primarily the matrix (M) gene with the conversion of U to C and A to G bases, high titers of antibodies to MV, and infiltration of B cells and T cells into the CNS. Neither the precipitating event nor biology underlying the MV infection is understood, nor is their any satisfactory treatment. We report the creation of a transgenic mouse model that mimics the cardinal features of SSPE. This was achieved by initially infecting mice expressing the MV receptor with lymphocytic choriomeningitis virus Cl 13, a virus that transiently suppressed their immune system. Infection by MV 10 days later resulted in persistent MV infection of neurons. Analysis of brains from infected mice showed the biased U to C hypermutations in the MV M gene and T and B lymphocyte infiltration. These sera contained high titers of antibodies to MV. Thus, a small animal model is now available to both molecularly probe the pathogenesis of SSPE and to test a variety of therapies to treat the disease.


Subject(s)
Lymphocytic choriomeningitis virus/immunology , Measles virus/immunology , Subacute Sclerosing Panencephalitis/immunology , Subacute Sclerosing Panencephalitis/virology , Animals , Base Sequence , Homeodomain Proteins/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Measles virus/genetics , Measles virus/pathogenicity , Membrane Cofactor Protein/genetics , Membrane Cofactor Protein/metabolism , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sequence Analysis, DNA
8.
J Virol ; 77(6): 3799-808, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12610154

ABSTRACT

CD4 and CD8 T lymphocytes infiltrate the parenchyma of mouse brains several weeks after intracerebral, intraperitoneal, or oral inoculation with the Chandler strain of mouse scrapie, a pattern not seen with inoculation of prion protein knockout (PrP(-/-)) mice. Associated with this cellular infiltration are expression of MHC class I and II molecules and elevation in levels of the T-cell chemokines, especially macrophage inflammatory protein 1beta, IFN-gamma-inducible protein 10, and RANTES. T cells were also found in the central nervous system (CNS) in five of six patients with Creutzfeldt-Jakob disease. T cells harvested from brains and spleens of scrapie-infected mice were analyzed using a newly identified mouse PrP (mPrP) peptide bearing the canonical binding motifs to major histocompatibility complex (MHC) class I H-2(b) or H-2(d) molecules, appropriate MHC class I tetramers made to include these peptides, and CD4 and CD8 T cells stimulated with 15-mer overlapping peptides covering the whole mPrP. Minimal to modest K(b) tetramer binding of mPrP amino acids (aa) 2 to 9, aa 152 to 160, and aa 232 to 241 was observed, but such tetramer-binding lymphocytes as well as CD4 and CD8 lymphocytes incubated with the full repertoire of mPrP peptides failed to synthesize intracellular gamma interferon (IFN-gamma) or tumor necrosis factor alpha (TNF-alpha) cytokines and were unable to lyse PrP(-/-) embryo fibroblasts or macrophages coated with (51)Cr-labeled mPrP peptide. These results suggest that the expression of PrP(sc) in the CNS is associated with release of chemokines and, as shown previously, cytokines that attract and retain PrP-activated T cells and, quite likely, bystander activated T cells that have migrated from the periphery into the CNS. However, these CD4 and CD8 T cells are defective in such an effector function(s) as IFN-gamma and TNF-alpha expression or release or lytic activity.


Subject(s)
Brain/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Creutzfeldt-Jakob Syndrome/immunology , Scrapie/immunology , Amino Acid Sequence , Animals , Cell Line , H-2 Antigens/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Peptides/chemistry , Peptides/metabolism , PrPSc Proteins/chemistry , PrPSc Proteins/metabolism
10.
J Virol ; 76(9): 4357-63, 2002 May.
Article in English | MEDLINE | ID: mdl-11932402

ABSTRACT

Transmissible spongiform encephalopathy or prion diseases are fatal neurodegenerative disorders of humans and animals often initiated by oral intake of an infectious agent. Current evidence suggests that infection occurs initially in the lymphoid tissues and subsequently in the central nervous system (CNS). The identity of infected lymphoid cells remains controversial, but recent studies point to the involvement of both follicular dendritic cells (FDC) and CD11c(+) lymphoid dendritic cells. FDC generation and maintenance in germinal centers is dependent on lymphotoxin alpha (LT-alpha) and LT-beta signaling components. We report here that by the oral route, LT-alpha -/- mice developed scrapie while LT-beta -/- mice did not. Furthermore, LT-alpha -/- mice had a higher incidence and shorter incubation period for developing disease following inoculation than did LT-beta -/- mice. Transplantation of lymphoid tissues from LT-beta -/- mice, which have cervical and mesenteric lymph nodes, into LT-alpha -/- mice, which do not, did not alter the incidence of CNS scrapie. In other studies, a virus that is tropic for and alters functions of CD11c(+) cells did not alter the kinetics of neuroinvasion of scrapie. Our results suggest that neither FDC nor CD11c(+) cells are essential for neuroinvasion after high doses of RML scrapie. Further, it is possible that an as yet unidentified cell found more abundantly in LT-alpha -/- than in LT-beta -/- mice may assist in the amplification of scrapie infection in the periphery and favor susceptibility to CNS disease following peripheral routes of infection.


Subject(s)
Genetic Predisposition to Disease , Lymphotoxin-alpha/genetics , Membrane Proteins/genetics , PrPSc Proteins/pathogenicity , Scrapie/physiopathology , Animals , Brain/metabolism , Dendritic Cells/immunology , Dendritic Cells, Follicular/immunology , Integrin alphaXbeta2/metabolism , Lymphoid Tissue/cytology , Lymphotoxin-beta , Mice , Mice, Inbred C57BL , Mice, Knockout , PrPSc Proteins/metabolism , Spleen/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...