Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Exp Med ; 217(12)2020 12 07.
Article in English | MEDLINE | ID: mdl-32880630

ABSTRACT

Intestinal barrier leakage constitutes a potential therapeutic target for many inflammatory diseases and represents a disease progression marker during chronic viral infections. However, the causes of altered gut barrier remain mostly unknown. Using murine infection with lymphocytic choriomeningitis virus, we demonstrate that, in contrast to an acute viral strain, a persistent viral isolate leads to long-term viral replication in hematopoietic and mesenchymal cells, but not epithelial cells (IECs), in the intestine. Viral persistence drove sustained intestinal epithelial barrier leakage, which was characterized by increased paracellular flux of small molecules and was associated with enhanced colitis susceptibility. Type I IFN signaling caused tight junction dysregulation in IECs, promoted gut microbiome shifts and enhanced intestinal CD8 T cell responses. Notably, both type I IFN receptor blockade and CD8 T cell depletion prevented infection-induced barrier leakage. Our study demonstrates that infection with a virus that persistently replicates in the intestinal mucosa increases epithelial barrier permeability and reveals type I IFNs and CD8 T cells as causative factors of intestinal leakage during chronic infections.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Interferon Type I/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/physiology , Animals , Antibodies/pharmacology , Chronic Disease , Clostridiales/physiology , Colitis/complications , Colitis/immunology , Colitis/virology , Epithelial Cells/virology , Female , Firmicutes , Gastrointestinal Microbiome , Gene Expression Regulation , Hematopoietic Stem Cells/virology , Intestinal Mucosa/microbiology , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/microbiology , Mesoderm/virology , Mice, Inbred C57BL , Permeability , Signal Transduction , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32958643

ABSTRACT

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Subject(s)
Anorexia/immunology , CD8 Antigens/immunology , Immunologic Memory/immunology , Lymphocytic Choriomeningitis/immunology , Virus Diseases/immunology , Akkermansia , Animals , Anorexia/microbiology , Anorexia/virology , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/virology , Firmicutes/immunology , Firmicutes/metabolism , Gastrointestinal Microbiome/immunology , Humans , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Verrucomicrobia/immunology , Verrucomicrobia/pathogenicity , Virus Diseases/microbiology , Virus Diseases/pathology
3.
Immunity ; 49(6): 1049-1061.e6, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566882

ABSTRACT

Appropriate immune responses require a fine balance between immune activation and attenuation. NLRC3, a non-inflammasome-forming member of the NLR innate immune receptor family, attenuates inflammation in myeloid cells and proliferation in epithelial cells. T lymphocytes express the highest amounts of Nlrc3 transcript where its physiologic relevance is unknown. We show that NLRC3 attenuated interferon-γ and TNF expression by CD4+ T cells and reduced T helper 1 (Th1) and Th17 cell proliferation. Nlrc3-/- mice exhibited increased and prolonged CD4+ T cell responses to lymphocytic choriomeningitis virus infection and worsened experimental autoimmune encephalomyelitis (EAE). These functions of NLRC3 were executed in a T-cell-intrinsic fashion: NLRC3 reduced K63-linked ubiquitination of TNF-receptor-associated factor 6 (TRAF6) to limit NF-κB activation, lowered phosphorylation of eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1), and diminished glycolysis and oxidative phosphorylation. This study reveals an unappreciated role for NLRC3 in attenuating CD4+ T cell signaling and metabolism.


Subject(s)
Autoimmunity/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Immunity, Innate/immunology , Intercellular Signaling Peptides and Proteins/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Adaptor Proteins, Signal Transducing , Animals , Autoimmunity/genetics , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Cycle Proteins , Encephalomyelitis, Autoimmune, Experimental/genetics , Eukaryotic Initiation Factors , Humans , Immunity, Innate/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphoproteins/genetics , Phosphoproteins/immunology , Phosphoproteins/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology , TNF Receptor-Associated Factor 6/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
4.
S Afr Med J ; 98(9): 724-5, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19113056

ABSTRACT

We retrospectively reviewed microbiological data from a tertiary care hospital in Botswana, and found that Cryptococcus neoformans was cultured from 15% (193/1307) of all cerebrospinal fluid (CSF) specimens submitted for analysis, making it the most common diagnosed cause of meningitis in this population. Moreover, almost 70% of CSF samples with significant lymphocytosis did not yield a pathogen, suggesting that many causes of lymphocytic meningitis go undiagnosed.


Subject(s)
Lymphocytic Choriomeningitis/epidemiology , Meningitis, Cryptococcal/epidemiology , Botswana/epidemiology , Cryptococcus neoformans/isolation & purification , Female , Humans , Lymphocytic Choriomeningitis/cerebrospinal fluid , Lymphocytic Choriomeningitis/microbiology , Male , Meningitis, Cryptococcal/cerebrospinal fluid , Meningitis, Cryptococcal/microbiology , Retrospective Studies
5.
J Immunol ; 171(11): 6032-8, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14634115

ABSTRACT

Memory CD8 T cells play a critical role in protective immunity against intracellular pathogens. In addition to their ability to specifically recognize and lyse infected targets, activated CD8 T cells secrete cytokines that induce phagocytic cells to engulf and kill bacterial pathogens. In this study, we asked whether activation of Ag-specific CD8 T cells results in nonspecific killing of bystander bacteria during a mixed infection. Mice with epitope-specific memory CD8 T cells were coinfected with two isogenic strains of recombinant Listeria monocytogenes that differ in the cognate epitope. Recall responses by epitope-specific CD8 T cells rapidly inhibited the growth of epitope-bearing bacteria, impeding the course of infection within 6 h after challenge. This rapid inhibition was highly specific and did not affect the growth of coinfecting bacteria without the epitope. CTL recall did not enhance activation of innate immune cells, as evidenced by the absence of inducible NO synthase production in infectious foci. Our observations demonstrate the remarkable specificity of the bactericidal mechanisms of CTL and reveal the possibility for escape mutants to prevail in the hostile environment of a specific immune response. This implication has a bearing on subunit vaccine design strategies and understanding failure of immunization against bacterial infection.


Subject(s)
Bystander Effect/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/microbiology , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/immunology , Listeria monocytogenes/growth & development , Listeria monocytogenes/immunology , Lymphocyte Activation/immunology , Amino Acid Sequence , Animals , CD8-Positive T-Lymphocytes/cytology , Female , Immunologic Memory , Listeria monocytogenes/genetics , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/pathology , Lymphocyte Count , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Macrophage Activation/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Nucleoproteins/administration & dosage , Nucleoproteins/genetics , Nucleoproteins/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
J Immunol ; 170(10): 4933-42, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12734336

ABSTRACT

Naive Ag-specific CD8(+) T cells expand, contract, and become memory cells after infection and/or vaccination. Memory CD8(+) T cells provide faster, more effective secondary responses against repeated exposure to the same pathogen. Using an adoptive transfer system with low numbers of trackable nontransgenic memory CD8(+) T cells, we showed that secondary responses can be comprised of both primary (naive) and secondary (memory) CD8(+) T cells after bacterial (Listeria monocytogenes) and/or viral (lymphocytic choriomeningitis virus) infections. The level of memory CD8(+) T cells present at the time of infection inversely correlated with the magnitude of primary CD8(+) T cell responses against the same epitope but directly correlated with the level of protection against infection. However, similar numbers of Ag-specific CD8(+) T cells were found 8 days postinfection no matter how many memory cells were present at the time of infection. Rapid contraction of primary CD8(+) T cell responses was not influenced by the presence of memory CD8(+) T cells. However, contraction of secondary CD8(+) T cell responses was markedly prolonged compared with primary responses in the same host mice. This situation occurred in response to lymphocytic choriomeningitis virus or L. monocytogenes infection and for CD8(+) T cell responses against multiple epitopes. The delayed contraction of secondary CD8(+) T cells was also observed after immunization with peptide-coated dendritic cells. Together, the results show that the level of memory CD8(+) T cells influences protective immunity and activation of naive precursors specific for the same epitope but has little impact on the magnitude or program of the CD8(+) T cell response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunization , Listeriosis/immunology , Lymphocytic Choriomeningitis/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/virology , Cell Division/immunology , Dendritic Cells/immunology , Dendritic Cells/transplantation , Epitopes, T-Lymphocyte/immunology , Immunization/methods , Immunization, Secondary/methods , Immunologic Memory , Interphase/immunology , Listeriosis/virology , Lymphocyte Activation , Lymphocyte Count , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/prevention & control , Mice , Mice, Inbred BALB C , Nucleoproteins/immunology , Peptide Fragments/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/microbiology , T-Lymphocyte Subsets/virology
7.
J Immunol ; 166(2): 982-8, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11145676

ABSTRACT

LPS is the major active agent in the pathogenesis of Gram-negative septic shock. In this report we have studied the influence of concurrent viral infection on the outcome of LPS-induced shock. We find that infection with vesicular stomatitis virus sensitizes mice to LPS at an early time point following infection. Treatment of mice with the chemical IFN inducer, polyinosinic:polycytidylic acid, has a similar effect. This hypersensitivity to LPS correlated with hyperproduction of TNF-alpha in vivo. The cellular and molecular mechanisms underlying this phenomenon were investigated using Ab-depleted and gene-targeted mice. Our results revealed that while NK cell depletion and elimination of IFN-gamma partially protected against the sensitizing effects of vesicular stomatitis virus and polyinosinic:polycytidylic acid, the most striking effect was observed in IFN-alphabetaR-deficient mice. Thus hyperproduction of TNF-alpha was completely abrogated in IFN-alphabetaR-deficient mice, indicating that the principal mechanism underlying rapid virus-induced sensitization to LPS is an IFN-alphabeta-mediated priming of mice for an augmented production of TNF-alpha in response to LPS. This conclusion was further supported by the finding that pretreatment of mice with rIFN-alphabeta mimicked the effect of viral infection. In conclusion, our results reveal a previously unrecognized proinflammatory effect of IFN-alphabeta and point to a new pathway through which viral infection may influence the outcome of concurrent bacterial infection.


Subject(s)
Interferon Type I/physiology , Lipopolysaccharides/toxicity , Rhabdoviridae Infections/immunology , Shock, Septic/immunology , Vesicular stomatitis Indiana virus/immunology , Animals , B-Lymphocytes/immunology , Disease Susceptibility , Female , Humans , Injections, Intraperitoneal , Interferon Inducers/administration & dosage , Interferon Type I/administration & dosage , Interferon-gamma/physiology , Killer Cells, Natural/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/mortality , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Poly I-C/administration & dosage , Recombinant Proteins , Rhabdoviridae Infections/microbiology , Rhabdoviridae Infections/mortality , Shock, Septic/mortality , Shock, Septic/virology , T-Lymphocytes/immunology , Time Factors , Tumor Necrosis Factor-alpha/biosynthesis
8.
J Neuroimmunol ; 71(1-2): 31-6, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8982100

ABSTRACT

To elucidate the possible involvement of the inducible nitric oxide synthase (iNOS) and NO in the development of lymphocytic choriomeningitis (LCM), the consequences of inhibition of iNOS by the inhibitor aminoguanidine was examined in mice following intracerebral infection with LCM virus (LCMV). Aminoguanidine administration to mice infected with LCMV completely blocked increased plasma nitrate/nitrite levels and led to increased proinflammatory cytokine gene expression at early stages of lesion development in the brain, enhanced clinical severity and decreased survival time. The levels of LCMV recovered from the brain of aminoguanidine treated mice did not differ from those in infected control mice. These findings argue against either an anti-viral or pathogenic role of NO in LCM but rather suggest a possible protective action of this mediator.


Subject(s)
Lymphocytic Choriomeningitis/physiopathology , Nitric Oxide Synthase/physiology , Animals , Brain/physiology , Cytokines/metabolism , Guanidines/pharmacology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus , Male , Mice , Mice, Inbred BALB C , Nitrates/blood , Nitric Oxide Synthase/antagonists & inhibitors , Nitrites/blood
9.
J Immunol ; 153(1): 220-31, 1994 Jul 01.
Article in English | MEDLINE | ID: mdl-8207238

ABSTRACT

IFN-alpha/beta have been shown to play a central role in the development of lymphocytic choriomeningitis and increasing attention has been focused on this group of cytokines as early regulatory factors directing T lymphocyte responses. In the present study, injection of antiserum to IFN-alpha/beta prevented the development of lymphocytic choriomeningitis, was associated with the absence of detectable expression of early 2'-5' oligo-adenylate synthetase mRNA and coincided with viremia of lymphocytic choriomeningitis virus (LCMV) followed by establishment of a persistent infection. The LCMV-specific cytotoxic T lymphocyte response was unchanged in cervical lymph nodes but decreased in the spleen of anti-IFN-alpha/beta-treated animals. The expression of cytokine mRNA (particularly IFN-gamma) in organs of LCMV-infected mice treated with anti-IFN-alpha/beta coincided with infiltration of lymphocytes and tissue destruction. Furthermore, a reduced number of infiltrating leukocytes in the brain and cervical lymph nodes and a low expression of cytokine mRNA in the brain was observed in anti-IFN-alpha/beta-treated animals. In total, the findings support the view that neutralization of IFN-alpha/beta leads to extensive LCMV replication in the viscera. The therapeutic effects of anti-IFN-alpha/beta antiserum seem to be independent of the functional capacity of T cells but probably result in a dispersion of activated T cells throughout the body of LCMV-infected mice. Absence of IFN-alpha/beta expression in the central nervous system is proposed as the mechanism behind the IFN-alpha/beta-dependent targeting of T cells to the brain.


Subject(s)
Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , T-Lymphocytes/microbiology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Antigen-Antibody Reactions , CD4-CD8 Ratio , Cell Adhesion , Cytokines/genetics , Cytotoxicity, Immunologic , Gene Expression , Interferon Type I/antagonists & inhibitors , Lymphocyte Activation , Lymphocytic Choriomeningitis/microbiology , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/growth & development , Male , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , T-Lymphocytes/cytology , Tissue Distribution , Virus Replication
10.
Virology ; 198(1): 1-9, 1994 Jan.
Article in English | MEDLINE | ID: mdl-8259643

ABSTRACT

The path that the arenavirus lymphocytic choriomeningitis virus (LCMV) uses to enter rodent fibroblastic cell lines was dissected by infectivity and inhibition studies and immunoelectron microscopy. Lysosomotropic weak bases (chloroquine and ammonium chloride) and carboxylic ionophores (monensin and nigericin) inhibited virus entry, assessed as virus nucleoprotein expression at early times post-infection, indicating that the entry process involved a pH-dependent fusion step in intracellular vesicles. That entry occurred in vesicles rather than by direct fusion of virions with the plasma membrane was confirmed by immunoelectron microscopy. The vesicles involved were large (150-300 nm diameter), smooth-walled, and not associated with clathrin. Unlike classical phagocytosis, virus uptake in these vesicles was a microfilament-independent process, as it was not blocked by cytochalasins. LCMV entry into rodent fibroblast cell lines thus involves viropexis in large smooth-walled vesicles, followed by a pH-dependent fusion event inside the cell.


Subject(s)
Lymphocytic choriomeningitis virus/pathogenicity , Alkalies/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Cell Membrane/metabolism , Cell Membrane/microbiology , Cricetinae , Fibroblasts/metabolism , Fibroblasts/microbiology , Hydrogen-Ion Concentration , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/drug effects , Lymphocytic choriomeningitis virus/ultrastructure , Lysosomes/metabolism , Lysosomes/microbiology , Mice , Receptors, Virus/drug effects , Receptors, Virus/physiology , Virus Replication/drug effects
11.
J Virol ; 67(12): 7340-9, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7693969

ABSTRACT

This study documents that the immunosuppressive lymphocytic choriomeningitis virus (LCMV) variant, clone 13, shows a specific predilection for enhanced infection of macrophages both in vitro and in vivo and that single amino acid changes in the viral polymerase and glycoprotein are responsible for macrophage tropism. The growth difference seen between variant clone 13 and the parental Armstrong strain was specific for macrophages, since both clone 13 and Armstrong grew equally well in fibroblasts and neither isolate infected lymphocytes efficiently. Complete sequencing of the clone 13 genome, along with genetic analysis, showed that a single amino acid change in the polymerase (K-->Q at position 1079) was the major determinant of virus yield in macrophages. This was proven unequivocally by comparing the sequences of parental and reassortant viruses, which were identical at all loci except for the single mutation in the polymerase gene. This finding was further strengthened by showing that reversion at this site back to lysine (Q-->K) resulted in loss of macrophage tropism. In addition, an independently derived macrophage-tropic variant of LCMV, clone 28b, had a K-->N mutation at the same position. Thus, these results show that substitution of the positively charged amino acid K with a neutral amino acid (either Q or N) at residue 1079 of the polymerase resulted in enhanced viral replication in macrophages. In addition to the polymerase change, a mutation in the glycoprotein was also associated with macrophage tropism. This single amino acid change in the glycoprotein (F-->L at position 260) did not affect virus yield per macrophage but was critical in determining the number of macrophages infected. Our previous studies have shown that the same two mutations in the polymerase and glycoprotein are essential for establishing a chronic infection in adult mice. Since the same mutations confer macrophage tropism and ability to persist in vivo, these studies provide compelling evidence that infection of macrophages is a critical determinant of viral persistence and immune suppression.


Subject(s)
Glycoproteins/genetics , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Macrophages/microbiology , RNA-Directed DNA Polymerase/genetics , Viral Proteins/genetics , Animals , Genetic Variation , Kupffer Cells/microbiology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Sequence Analysis , Spleen/microbiology , Virulence , Virus Replication
12.
Nature ; 362(6422): 758-61, 1993 Apr 22.
Article in English | MEDLINE | ID: mdl-8469287

ABSTRACT

Viruses that are non- or poorly cytopathic have developed various strategies to avoid elimination by the immune system and to persist in the host. Acute infection of adult mice with the noncytopathic lymphocytic choriomeningitis virus (LCMV) normally induces a protective cytotoxic T-cell response that also causes immunopathology. But some LCMV strains (such as DOCILE (LCMV-D) or Cl-13 Armstrong (Cl-13)) derived from virus carrier mice tend to persist after acute infection of adult mice without causing lethal immunopathological disease. Tendency to persist correlates with tropism, rapidity of virus spread and virus mutations. We report here that these LCMV isolates may persist because they induce most of the specific antiviral CD8+ cytotoxic T cells so completely that they all disappear within a few days and therefore neither eliminate the virus nor cause lethal immunopathology. The results illustrate that partially and sequentially induced (protective) immunity or complete exhaustion of T-cell immunity (high zone tolerance) are quantitatively different points on the scale of immunity; some viruses exploit the latter possibility to persist in an immunocompetent host.


Subject(s)
Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , CD8 Antigens , Immune Tolerance , Immunocompetence , Immunophenotyping , Immunotherapy, Adoptive , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Spleen/immunology , T-Lymphocytes, Cytotoxic/transplantation
13.
Intervirology ; 35(1-4): 116-21, 1993.
Article in English | MEDLINE | ID: mdl-8407238

ABSTRACT

Viral persistence depends on a virus having a non-lytic strategy of replication and the ability to escape immune surveillance. Cells of the immune system (lymphocytes/monocytes/macrophages) and central nervous system (neurons) are most often infected by DNA and RNA viruses that persist. Cytotoxic T lymphocytes (CTL) are the primary host defense that aborts or prevents viral persistence. Viral interaction with these specialized cells and of such infected cells with CTL is explored in this paper.


Subject(s)
Immune System/microbiology , Neurons/microbiology , Animals , Immune System/cytology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/physiology , T-Lymphocytes, Cytotoxic/microbiology , Virus Replication
14.
JAMA ; 267(10): 1349-53, 1992 Mar 11.
Article in English | MEDLINE | ID: mdl-1740856

ABSTRACT

OBJECTIVE: After an employee at a cancer research institute was diagnosed with lymphocytic choriomeningitis, an investigation was performed to determine the extent of lymphocytic choriomeningitis virus (LCMV) infections among the institute's employees and to identify risk factors for infection. DESIGN: Retrospective cohort study. SETTING: A US cancer research institute. PARTICIPANTS: Eighty-two of 90 institute employees. MAIN OUTCOME MEASURES: Serum LCMV antibodies. RESULTS: Seven workers (9%) with definite LCMV infection (LCMV IgG antibody titer greater than or equal to 16) and one worker (1%) with probable infection (IgG titer = 8) were identified (10% overall seroprevalence). All infected employees handled animals or animal tissues and were more likely than other animal handlers to have worked with nude mice (Mus musculus) (P less than .02). Among the 31 employees who worked with nude mice at the institute, infected workers were more likely to clean the cages of nude mice (P much less than .001), change their bedding (P less than .01), and change their water (P less than .001). The institute had been injecting nude mice with LCMV-infected tumor cell lines and had recently increased the nude mouse population and the duration of experiments. These changes would have increased the LCMV burden at the facility and were temporally associated with the cluster of LCMV infections in employees. CONCLUSIONS: This LCMV outbreak, the first reported since 1974, is the first associated with nude mice. It illustrates the ongoing hazard LCMV poses in research laboratories. Since the symptoms of LCMV infection can be nonspecific, clinicians should consider this diagnosis in ill patients who report laboratory rodent exposure.


Subject(s)
Disease Outbreaks , Laboratory Infection/epidemiology , Lymphocytic Choriomeningitis/epidemiology , Mice, Nude/microbiology , Adult , Animals , Antibodies, Viral/analysis , Female , Humans , Laboratory Infection/immunology , Laboratory Infection/microbiology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Nude/immunology , Rodent Diseases/immunology , Rodent Diseases/microbiology , Rodent Diseases/transmission
16.
J Immunol ; 147(9): 3133-8, 1991 Nov 01.
Article in English | MEDLINE | ID: mdl-1919008

ABSTRACT

In (B10.BR----B10) chimeras infected with lymphocytic choriomeningitis (LCM) virus higher titers were attained in spleens and livers than in organs of the mice used for their construction, and the subsequent elimination was retarded, but eventually the virus was cleared. The numbers of LCM virus-specific CTL and their precursors as quantitated with chromium-release assay and limiting dilution method, respectively, were lower in chimeras than in B10.BR or C57BL/10J mice, and fewer were restricted for the haplotypes of the donors than of the recipients. The same was true with regard to antiviral effector cells, which were determined by adoptive immunization. The numbers of spleen cells releasing IgM and IgG antiviral antibodies were virtually as high in chimeras as they were in C57BL/10J and B10.BR mice. Transfer of immune splenocytes from either B10.BR or C57BL/10J mice resulted in incomplete virus elimination from the spleens of infected chimeras, whereas injection of a mixture of the two types of immune cells led to efficient clearance. We conclude that in the chimeras cells of both donor and recipient haplotypes participate in the infection, which is terminated by H-2k- and H-2b-restricted T lymphocytes that these animals are capable of generating. We conclude, furthermore, that clearance of the LCM virus from the tissues requires contact between effector and target cells.


Subject(s)
Bone Marrow/immunology , Lymphocytic Choriomeningitis/immunology , Radiation Chimera , Animals , Antibodies, Viral/biosynthesis , Antibody Formation , Bone Marrow Transplantation/immunology , Cytotoxicity, Immunologic , H-2 Antigens/immunology , Immunity, Cellular , Immunization, Passive , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred Strains , Spleen/cytology , Spleen/microbiology , T-Lymphocytes, Cytotoxic/immunology
17.
J Vet Med Sci ; 53(5): 889-92, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1836377

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) was isolated from 14 out of 35 wild house mouse samples captured on two piers of Osaka port in Japan. Four of them were isolated from were isolated from 18 antibody positive mice while 10 were from 17 antibody negative mice. This is the first report of the isolation of LCMV from wild mice in Japan.


Subject(s)
Animals, Wild , Lymphocytic Choriomeningitis/veterinary , Lymphocytic choriomeningitis virus/isolation & purification , Mice , Rodent Diseases/epidemiology , Age Factors , Animals , Antibodies, Viral/blood , Disease Reservoirs , Japan/epidemiology , Lymphocytic Choriomeningitis/epidemiology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred BALB C , Prevalence , Rodent Diseases/microbiology , Vero Cells
18.
J Virol ; 65(8): 4242-7, 1991 Aug.
Article in English | MEDLINE | ID: mdl-2072451

ABSTRACT

Viral variants of different phenotypes are present in the central nervous system (CNS) and lymphoid tissues of carrier mice infected at birth with the Armstrong strain of lymphocytic choriomeningitis virus. The CNS isolates are similar to the parental virus and cause acute infections in adult mice, whereas the lymphoid isolates cause chronic infections associated with suppressed T-cell responses. In this study, we provide a molecular basis for this organ-specific selection and identify a single amino acid change in the viral glycoprotein that correlates with the tissue specific selection and the persistent and immunosuppressive phenotype of the variants. This phenylalanine (F)-to-leucine (L) change at position 260 of the viral glycoprotein was seen in the vast majority (43 of 47) of the lymphoid isolates, and variants with L at this residue were selected in spleens of persistently infected mice. In striking contrast, isolates with the parental sequence (F at residue 260) predominated (48 of 59 isolates) in the CNS of the same carrier mice. Complete nucleotide sequence analysis of the major structural genes of several independently derived (from different mice) spleen isolates showed that these variants were greater than 99.8% identical to the parental virus. In fact, the only common change among these spleen isolates was the F----L mutation at residue 260 of the glycoprotein. These results show that an RNA virus can exhibit minimal genetic drift during chronic infection in its natural host, and yet a single or few mutations can result in the organ-specific selection of variants that are markedly different from the parental virus.


Subject(s)
Brain/microbiology , Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/physiology , Spleen/microbiology , Amino Acid Sequence , Animals , Base Sequence , Carrier State/microbiology , Chronic Disease , Gene Frequency , Glycoproteins/chemistry , Glycoproteins/genetics , Leucine , Lymphocytic choriomeningitis virus/genetics , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Mutation , Oligonucleotides/chemistry , Organ Specificity , Phenylalanine , RNA, Viral/chemistry , Viral Proteins/chemistry , Viral Proteins/genetics
19.
J Gen Virol ; 72 ( Pt 7): 1611-25, 1991 Jul.
Article in English | MEDLINE | ID: mdl-1649899

ABSTRACT

By the application of in situ hybridization to thin sections of paraffin-embedded tissues we have been able to determine with high resolution the cell types containing lymphocytic choriomeningitis virus nucleic acid in the tissues of persistently infected mice. We confirm and extend previous observations of virus persistence in the brain, lung, liver, kidney, pancreas, thyroid and reticuloendothelial system. In addition, we demonstrate for the first time persistence of viral nucleic acid in specific cell types in the thymus, lymph nodes, testes and bladder, and the adrenal, parathyroid and salivary glands; the cell types infected were observed in several animals. In lymphoid tissue, viral nucleic acid was predominantly located in the T cell-dependent areas of the spleen and lymph nodes; it was also present in cells of the thymic medulla. This has important implications for the deficiency in T cell function observed in persistently infected mice. In the testes, viral nucleic acid was detected in spermatogonia but not differentiating spermatocytes and therefore, in this tissue at least, persistence is related to the state of differentiation of the cell. Endocrine and exocrine dysfunctions have been described in persistently infected mice and we report that the highest levels of viral nucleic acid were found in the adrenal gland. The infection of endocrine and exocrine tissue was not pantropic, specific cell types expressed viral nucleic acid in each tissue. In the adrenal cortex, cells of the zona reticularis and zona fasciculata but not the zona glomerulosa were positive, whereas in the adrenal medulla viral nucleic acid was predominantly localized to adrenalin-secreting cells. Infection of the renal tubules, transitional epithelium of the bladder and the ducts of the salivary gland indicates the likely sites of virus production for the dissemination of arenavirus infections.


Subject(s)
Lymphocytic Choriomeningitis/microbiology , Lymphocytic choriomeningitis virus/genetics , RNA, Viral/analysis , Adrenal Glands/microbiology , Animals , Arenaviridae Infections/microbiology , Brain/microbiology , Lymphocytic choriomeningitis virus/physiology , Male , Mice , Mice, Inbred BALB C , Mononuclear Phagocyte System/microbiology , Neurons/microbiology , Nucleic Acid Hybridization , Pancreas/microbiology , Parathyroid Glands/microbiology , Salivary Glands/microbiology , Testis/microbiology , Thymus Gland/microbiology
20.
Adv Pediatr Infect Dis ; 6: 111-36, 1991.
Article in English | MEDLINE | ID: mdl-1647166

ABSTRACT

Nonherpetic encephalitis outside the newborn period is usually a self-limited disease. The majority of patients will recover without significant sequelae, and require only supportive therapy during the acute illness. Though the underlying viral etiology frequently will escape detection, identification of the infecting agent has considerable prognostic value which can complement clinical measures of severity of disease. The most important initial task of the clinician faced with a case of presumptive viral encephalitis is to eliminate the possibility of a treatable illness. Once this has been done, the diagnosis of viral encephalitis can be supported by documenting the characteristic slow-wave background activity on EEG, and a mild lymphocellular pleocytosis in the CSF. Because viral encephalitis can be caused by such a large number of organisms, the search for an etiology can be daunting. Realizing that all the agents described above can, at times, cause encephalitis without any clue to their identity, one nevertheless may use several pieces of historical information to narrow the possibilities. Travel history, animal exposures, immunization history, and seasonality all may help to steer the search in a particular direction and, indeed, may point to a nonvirologic cause as well. In addition, detection of extraneurological signs and symptoms may strongly indicate a specific virologic diagnosis. Finally, knowledge of concurrent community epidemic patterns, and of surveillance data routinely collected by local and state health departments, can help to increase or decrease the likelihood of a given pathogen. The causative viral agent usually can be identified by serological testing and viral culture. Occasionally, single serological determinations are diagnostic: in rabies (when the patient has not received immune prophylaxis), eastern equine encephalitis, and HIV, since seropositivity is strongly associated with symptomatic illness; and in Epstein-Barr virus, if a panel of antibody determinations which can time the infection is available. In addition, high CSF: serum titers for antibody against any neurotropic agent is usually diagnostic, though the absence of a high central nervous system antibody titer does not eliminate any potential viral pathogen. With these few exceptions, a single serological determination for a given pathogen is almost always impossible to interpret; paired sera (one obtained upon diagnosis, and one obtained 10 to 14 days later, either just prior to hospital discharge or at a follow-up visit) are far more helpful. Many viruses that directly infect the central nervous system are difficult to recover from the CSF; therefore, viral isolation from the nasopharynx and stool also should be sought.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Encephalitis/microbiology , Herpes Simplex/microbiology , Adenoviridae Infections/microbiology , Arbovirus Infections/microbiology , Diagnosis, Differential , Encephalitis Virus, Eastern Equine/isolation & purification , Encephalomyelitis, Equine/microbiology , Enterovirus Infections/microbiology , HIV Infections/microbiology , Herpesviridae Infections/microbiology , Humans , Lymphocytic Choriomeningitis/microbiology , Measles/microbiology , Rabies/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...