Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Int J Infect Dis ; 105: 452-459, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33667697

ABSTRACT

OBJECTIVES: Lymphocytic choriomeningitis virus (LCMV), a human pathogenic arenavirus, is distributed worldwide. However, no human cases have been reported in Africa. This study aimed to investigate the current situation and potential risks of LCMV infection in Gabon, Central Africa. METHODS: A total of 492 human samples were screened to detect LCMV genome RNA and anti-LCMV IgG antibodies using reverse transcription-quantitative PCR and enzyme-linked immunosorbent assay (ELISA), respectively. ELISA-positive samples were further examined using a neutralization assay. Viral RNAs and antibodies were also analyzed in 326 animal samples, including rodents, shrews, and bushmeat. RESULTS: While no LCMV RNA was detected in human samples, the overall seroprevalence was 21.5% and was significantly higher in male and adult populations. The neutralization assay identified seven samples with neutralizing activity. LCMV RNA was detected in one species of rodent (Lophuromys sikapusi) and a porcupine, and anti-LCMV IgG antibodies were detected in four rodents and three shrews. CONCLUSIONS: This study determined for the first time the seroprevalence of LCMV in Gabon, and revealed that local rodents, shrews, and porcupines in areas surrounding semi-urban cities posed an infection risk. Hence, LCMV infection should be considered a significant public health concern in Africa.


Subject(s)
Lymphocytic Choriomeningitis/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antibodies, Viral/blood , Child , Child, Preschool , Female , Gabon/epidemiology , Humans , Infant , Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Male , Middle Aged , RNA, Viral/blood , Seroepidemiologic Studies , Shrews , Young Adult
2.
Transpl Infect Dis ; 19(4)2017 Aug.
Article in English | MEDLINE | ID: mdl-28423464

ABSTRACT

Despite careful donor screening, unexpected donor-derived infections continue to occur in organ transplant recipients (OTRs). Lymphocytic choriomeningitis virus (LCMV) is one such transplant-transmitted infection that in previous reports has resulted in a high mortality among the affected OTRs. We report a LCMV case cluster that occurred 3 weeks post-transplant in three OTRs who received allografts from a common organ donor in March 2013. Following confirmation of LCMV infection at Centers for Disease Control and Prevention, immunosuppression was promptly reduced and ribavirin and/or intravenous immunoglobulin therapy were initiated in OTRs. The liver recipient died, but right kidney recipients survived without significant sequelae and left kidney recipient survived acute LCMV infection with residual mental status deficit. Our series highlights how early recognition led to prompt therapeutic intervention, which may have contributed to more favorable outcome in the kidney transplant recipients.


Subject(s)
Kidney Transplantation/adverse effects , Liver Transplantation/adverse effects , Lymphocytic Choriomeningitis/diagnosis , Lymphocytic choriomeningitis virus/isolation & purification , Aged , Donor Selection , Early Medical Intervention , Fatal Outcome , Female , Humans , Immunosuppression Therapy , Kidney/pathology , Kidney/virology , Liver/pathology , Liver/virology , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Male , Middle Aged , Tissue Donors , Transplant Recipients , Transplantation, Homologous
3.
Occup Med (Lond) ; 66(2): 171-3, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26416845

ABSTRACT

We report a case of acquired lymphocytic choriomeningitis virus (LCMV) infection due to an accidental percutaneous inoculation of LCMV at work. The injured worker developed a flu-like syndrome, followed by pericarditis and meningoencephalitis. Seroconversion was confirmed by ELISA. The patient made a complete recovery. We review measures undertaken to prevent a similar event and propose a follow-up protocol in the event of accidental LCMV exposure.


Subject(s)
Accidents, Occupational , Antiviral Agents/administration & dosage , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic choriomeningitis virus/pathogenicity , Needlestick Injuries/virology , Occupational Exposure/adverse effects , Ribavirin/administration & dosage , Adult , Humans , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/virology , Male , Post-Exposure Prophylaxis , Practice Guidelines as Topic , Treatment Outcome
4.
J Immunol ; 195(1): 116-25, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25980007

ABSTRACT

Patients who survive sepsis display suppressed immune functions, often manifested as an increased susceptibility to secondary infections. Recently, using a cecal-ligation and puncture (CLP) model of sepsis, we showed that sepsis induces substantial and long-lasting changes in the available naive CD8(+) T cell repertoire affecting the capacity of the host to respond to newly encountered acute infections. However, the extent to which sepsis changes the host susceptibility to chronic infection and affects CD8(+) T cell responses is currently unknown. In this study, we demonstrate that inbred and outbred mice recovering from a septic event are more susceptible to lymphocytic choriomeningitis virus (LCMV) clone-13 infection exhibited by mortality and viral burden. Primary virus-specific CD8(+) T cells in LCMV clone-13-infected septic mice displayed exacerbated CD8(+) T cell exhaustion illustrated by increased inhibitory molecule expression (e.g., programmed cell death 1, lymphocyte-activation gene 3, and 2B4) and diminished Ag-driven cytokine production (e.g., IFN-γ, TNF-α) compared with similarly infected sham-treated mice. Importantly, therapeutic inhibitory molecule dual blockade (anti-PD-L1 and anti-lymphocyte-activation gene 3) increased the number of circulating LCMV-specific CD8(+) T cells, and improved CD8(+) T cell function and pathogen control in chronically infected septic mice. Together, these results illustrate that polymicrobial sepsis compromises the overall health of the host leading to increased vulnerability to chronic infection and exacerbated CD8(+) T cell exhaustion. Collectively, our findings suggest that septic survivors may be more susceptible and at greater risk for developing exhaustible CD8(+) T cells upon encountering a subsequent chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Host-Pathogen Interactions/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Sepsis/immunology , Animals , Antibodies/pharmacology , Antigens, CD/genetics , Antigens, CD/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/pathology , Convalescence , Disease Susceptibility , Gene Expression Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Count , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/mortality , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Primary Cell Culture , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Sepsis/complications , Sepsis/mortality , Sepsis/pathology , Signal Transduction , Signaling Lymphocytic Activation Molecule Family , Survival Analysis , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Viral Load , Lymphocyte Activation Gene 3 Protein
5.
Semin Pediatr Neurol ; 19(3): 89-95, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22889536

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV) is an important cause of neurologic disease in humans. Carried and secreted principally by wild mice, LCMV covers a large geographic range and infects great numbers of people. Humans acquire LCMV disease when they come into contact with the secretions of infected mice. Because it has a strong neurotropism, the clinical signs and symptoms of LCMV infection are mostly neurologic. When the virus is acquired postnatally by children or adults, the clinical manifestations are usually those of aseptic meningitis. Most people who acquire LCMV infection during childhood or adulthood are moderately symptomatic for several weeks, but have a full recovery. A much more severe disease ensues when the infection occurs prenatally. LCMV can infect the fetal brain and retina, where it leads to substantial injury and permanent dysfunction. The possibility of LCMV infection should be considered in all babies with evidence of congenital infection, especially those with prominent neurologic signs, such as microencephaly, periventricular calcifications, and hydrocephalus.


Subject(s)
Brain Diseases/virology , Fetal Diseases/virology , Lymphocytic Choriomeningitis , Lymphocytic choriomeningitis virus/pathogenicity , Animals , Antiviral Agents/therapeutic use , Brain Diseases/pathology , Diagnosis, Differential , Fetal Diseases/diagnosis , Humans , Lymphocytic Choriomeningitis/diagnosis , Lymphocytic Choriomeningitis/epidemiology , Lymphocytic Choriomeningitis/etiology , Prognosis
7.
J Pediatr Hematol Oncol ; 30(10): 781-4, 2008 Oct.
Article in English | MEDLINE | ID: mdl-19011481

ABSTRACT

We report a case of 15-year-old girl with T-cell acute lymphoblastic leukemia who had fever, neutropenia, and severe headache while receiving maintenance chemotherapy. Cerebrospinal fluid testing revealed a lymphocytic pleocytosis and no evidence of relapsed leukemia. Meningitis caused by lymphocytic choriomeningitis virus was identified serologically. The patient's course was complicated by hydrocephalus requiring ventriculoperitoneal shunt placement and by an intracranial hemorrhage. Lymphocytic choriomeningitis virus is a rare cause of aseptic meningitis that should be considered in the symptomatic immunocompromised patient with an appropriate exposure history.


Subject(s)
Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/isolation & purification , Meningitis, Aseptic/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Adolescent , Female , Humans , Hydrocephalus/etiology , Immunocompromised Host , Intracranial Hemorrhages/etiology , Lymphocytic Choriomeningitis/virology , Meningitis, Aseptic/virology , Serologic Tests
8.
J Virol ; 80(3): 1222-30, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16414999

ABSTRACT

Intracerebral (i.c.) infection with lymphocytic choriomeningitis virus (LCMV) is one of the most studied models for virus-induced immunopathology, and based on results from perforin-deficient mice, it is currently assumed that fatal disease directly reflects perforin-mediated cell lysis. However, recent studies have revealed additional functional defects within the effector T cells of LCMV-infected perforin-deficient mice, raising the possibility that perforin may not be directly involved in mediating lethal disease. For this reason, we decided to reevaluate the role of perforin in determining the outcome of i.c. infection with LCMV. We confirmed that the expansion of virus-specific CD8(+) T cells is unimpaired in perforin-deficient mice. However, despite the fact that the virus-specific CD8(+) effector T cells in perforin-deficient mice are broadly impaired in their effector function, these mice invariably succumb to i.c. infection with LCMV strain Armstrong, although a few days later than matched wild-type mice. Upon further investigation, we found that this delay correlates with the delayed recruitment of inflammatory cells to the central nervous system (CNS). However, CD8(+) effector T cells were not kept from the CNS by sequestering in infected extraneural organ sites such as liver or lungs. Thus, the observed dysfunctionality regarding the production of proinflammatory mediators probably results in the delayed recruitment of effector cells to the CNS, and this appears to be the main explanation for the delayed onset of fatal disease in perforin-deficient mice. However, once accumulated in the CNS, virus-specific CD8(+) T cells can induce fatal CNS pathology despite the absence of perforin-mediated lysis and reduced capacity to produce several key cytokines.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/biosynthesis , Lymphocytic Choriomeningitis/immunology , Membrane Proteins/deficiency , Animals , Disease Models, Animal , Inflammation Mediators/metabolism , Integrin alpha4beta1/biosynthesis , Interferon-gamma/biosynthesis , Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/pathogenicity , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pore Forming Cytotoxic Proteins
9.
Ir Med J ; 98(10): 238-40, 2005.
Article in English | MEDLINE | ID: mdl-16445142

ABSTRACT

Over a five-year period 114 cases of lymphocytic meningitis were admitted to a general hospital. Case notes were reviewed to determine incidence, causes and outcomes and to identify what clinical features, cerebrospinal fluid (CSF) parameters or other tests were useful in elucidating a cause. Of 114 patients with lymphocytic meningitis 44 had viral meningitis, six viral encephalitis, and seven had other infective meningo-encephalitis. Seven patients had carcinomatous meningitis, nine had an autoimmune/inflammatory process, thirteen had a demyelinating central nervous system (CNS) disorder, and two had an inflammatory neuropathy. In 26 patients no diagnosis was reached. Six of these showed clear steroid responsiveness. Overall lymphocytic meningitis had a good prognosis. Clinical and CSF characteristics were important in diagnosis and prognosis. Recommendations on management of these cases are presented.


Subject(s)
Lymphocytic Choriomeningitis/epidemiology , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/therapy , Outcome Assessment, Health Care , Adult , Diagnosis, Differential , Female , Humans , Incidence , Ireland/epidemiology , Male , Middle Aged , Retrospective Studies
10.
J Immunol ; 173(8): 4779-90, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15470017

ABSTRACT

Although both self- and pathogen-specific T cells can participate in tissue destruction, recent studies have proposed that after viral infection, bystander T cells of an irrelevant specificity can bypass peptide-MHC restriction and contribute to undesired immunopathological consequences. To evaluate the importance of this mechanism of immunopathogenesis, we determined the relative contributions of Ag-specific and bystander CD8+ T cells to the development of CNS disease. Using lymphocytic choriomeningitis virus (LCMV) as a stimulus for T cell recruitment into the CNS, we demonstrate that bystander CD8+ T cells with an activated surface phenotype can indeed be recruited into the CNS over a chronic time window. These cells become anatomically positioned in the CNS parenchyma, and a fraction aberrantly acquires the capacity to produce the effector cytokine, IFN-gamma. However, when directly compared with their virus-specific counterparts, the contribution of bystander T cells to CNS damage was insignificant in nature (even when specifically activated). Although bystander T cells alone failed to cause tissue injury, transferring as few as 1000 naive LCMV-specific CD8+ T cells into a restricted repertoire containing only bystander T cells was sufficient to induce immune-mediated pathology and reconstitute a fatal CNS disease. These studies underscore the importance of specific T cells in the development of immunopathology and subsequent disease. Because of highly restrictive constraints imposed by the host, it is more likely that specific, rather than nonspecific, bystander T cells are the active participants in T cell-mediated diseases that afflict humans.


Subject(s)
Brain/immunology , H-2 Antigens/immunology , Lymphocytic Choriomeningitis/immunology , Peptide Fragments/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Apoptosis , Brain/pathology , Cell Movement , Cytokines/biosynthesis , Histocompatibility Antigen H-2D , Hyaluronan Receptors/analysis , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/pathology , Mice , Mice, Inbred C57BL , Molecular Sequence Data
11.
Arch Neurol ; 61(10): 1553-7, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15477509

ABSTRACT

BACKGROUND: Herpes simplex virus 2 (HSV-2) and HSV-1 have been recognized as causes of recurrent aseptic lymphocytic meningitis (RALM). However, the role of other herpesviruses has not been systematically assessed. OBJECTIVES: To evaluate the cause of RALM by using polymerase chain reaction (PCR) tests detecting varicella-zoster virus (VZV), cytomegalovirus (CMV), or human herpesvirus 6 (HHV-6), in addition to HSV, on cerebrospinal fluid (CSF) samples; and to assess the utility of PCR and antibody analyses in consecutive episodes of RALM. DESIGN: The PCR and antibody results for herpesviruses were analyzed from 14 patients having 48 episodes of RALM. RESULTS: The CSF PCR results for VZV, CMV, and HHV-6 were negative in 12, 10, and 11 patients investigated, respectively, and antibodies against VZV, CMV, and HHV-6 showed only old immunity. Herpes simplex virus 2 was detected from the CSF in 10 patients, and HSV-1 in 1 patient. In 6 of these 11 patients, the HSV PCR result was positive in more than one disease episode. A significant increase of serum antibodies for HSV was seen in only 1 of 15 episodes examined. An intrathecal antibody response to HSV was not recognized in 9 episodes investigated in these 11 patients. CONCLUSIONS: We could not find evidence of VZV, CMV, or HHV-6 in the pathogenesis of RALM, although most patients were previously infected by those viruses. Herpes simplex virus 2 was detected from the CSF in most patients, and often repeatedly, which further confirms the role of this virus in RALM. The causative diagnosis was obtained only by PCR, whereas antibody analysis was not clinically useful.


Subject(s)
Herpesviridae Infections/complications , Herpesvirus 6, Human/pathogenicity , Lymphocytic Choriomeningitis/virology , Adult , Cerebrospinal Fluid/virology , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/cerebrospinal fluid , Cytomegalovirus Infections/etiology , Cytomegalovirus Infections/virology , Female , Herpesviridae Infections/cerebrospinal fluid , Herpesviridae Infections/virology , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/isolation & purification , Herpesvirus 3, Human/pathogenicity , Herpesvirus 6, Human/genetics , Herpesvirus 6, Human/isolation & purification , Humans , Lymphocytic Choriomeningitis/cerebrospinal fluid , Lymphocytic Choriomeningitis/etiology , Male , Middle Aged , Polymerase Chain Reaction/methods , Retrospective Studies , Statistics, Nonparametric
12.
Virology ; 316(2): 213-20, 2003 Nov 25.
Article in English | MEDLINE | ID: mdl-14644604

ABSTRACT

Dystroglycan (DG) is a highly versatile cell surface molecule that provides a molecular link between the extracellular matrix (ECM) and the actin-based cytoskeleton. Encoded by a single gene, DG is posttranslationally processed to form alpha-DG, a peripheral protein identified as the cellular receptor for lymphocytic choriomeningitis virus (LCMV) and Lassa fever virus (LFV), and the membrane-spanning subunit beta-DG. The link of beta-DG to the actin-based cytoskeleton and its association with the cellular signal transduction network suggest that it may function as an essential cofactor for the activity of alpha-DG as a virus receptor. To address this issue, we constructed a deletion mutant lacking the cytoplasmic domain of beta-DG and a C-terminal fusion between alpha-DG and the transmembrane domain of PDGF receptor. Both mutants were functional as virus receptors, indicating that beta-DG does not act as a cofactor with alpha-DG for arenavirus binding and entry. These observations are in agreement with the fact that LCMV infection is independent from the structural integrity of the actin-based cytoskeleton and suggest that alpha-DG functions primarily in the attachment of arenaviruses to the cell surface.


Subject(s)
Cytoskeletal Proteins/physiology , Lymphocytic Choriomeningitis/etiology , Membrane Glycoproteins/physiology , Receptors, Virus/physiology , Animals , Cricetinae , Dystroglycans
13.
J Virol ; 76(16): 8347-59, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12134039

ABSTRACT

Hantavirus cardiopulmonary syndrome (HCPS) is a life-threatening respiratory disease characterized by profound pulmonary edema and myocardial depression. Most cases of HCPS in North America are caused by Sin Nombre virus (SNV), which is carried asymptomatically by deer mice (Peromyscus maniculatus). The underlying pathophysiology of HCPS is poorly understood. We hypothesized that pathogenic SNV infection results in increased generation of reactive oxygen/nitrogen species (RONS), which contribute to the morbidity and mortality of HCPS. Human disease following infection with SNV or Andes virus was associated with increased nitrotyrosine (NT) adduct formation in the lungs, heart, and plasma and increased expression of inducible nitric oxide synthase (iNOS) in the lungs compared to the results obtained for normal human volunteers. In contrast, NT formation was not increased in the lungs or cardiac tissue from SNV-infected deer mice, even at the time of peak viral antigen expression. In a murine (Mus musculus) model of HCPS (infection of NZB/BLNJ mice with lymphocytic choriomeningitis virus clone 13), HCPS-like disease was associated with elevated expression of iNOS in the lungs and NT formation in plasma, cardiac tissue, and the lungs. In this model, intraperitoneal injection of 1400W, a specific iNOS inhibitor, every 12 h during infection significantly improved survival without affecting intrapulmonary fluid accumulation or viral replication, suggesting that cardiac damage may instead be the cause of mortality. These data indicate that elevated production of RONS is a feature of pathogenic New World hantavirus infection and that pharmacologic blockade of iNOS activity may be of therapeutic benefit in HCPS cases, possibly by ameliorating the myocardial suppressant effects of RONS.


Subject(s)
Hantavirus Pulmonary Syndrome/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Tyrosine/analogs & derivatives , Adolescent , Adult , Amidines/pharmacology , Animals , Benzylamines/pharmacology , Blood Proteins/metabolism , Case-Control Studies , Disease Models, Animal , Disease Vectors , Enzyme Inhibitors/pharmacology , Female , Orthohantavirus/pathogenicity , Hantavirus Pulmonary Syndrome/etiology , Hantavirus Pulmonary Syndrome/pathology , Humans , Lymphocytic Choriomeningitis/drug therapy , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/metabolism , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred NZB , Middle Aged , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Peromyscus/virology , Sin Nombre virus/pathogenicity , Tyrosine/metabolism
14.
J Virol ; 75(1): 448-57, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119613

ABSTRACT

alpha-Dystroglycan (alpha-DG) was recently identified as a receptor for lymphocytic choriomeningitis virus (LCMV) and several other arenaviruses, including Lassa fever virus (W. Cao, M. D. Henry, P. Borrow, H. Yamada, J. H. Elder, E. V. Ravkov, S. T. Nichol, R. W. Compans, K. P. Campbell, and M. B. A. Oldstone, Science 282:2079-2081, 1998). Data presented in this paper indicate that the affinity of binding of LCMV to alpha-DG determines viral tropism and the outcome of infection in mice. To characterize this relationship, we evaluated the interaction between alpha-DG and several LCMV strains, variants, and reassortants. These viruses could be divided into two groups with respect to affinity of binding to alpha-DG, dependence on this protein for cell entry, viral tropism, and disease course. Viruses that exhibited high-affinity binding to alpha-DG displayed a marked dependence on alpha-DG for cell entry and were blocked from infecting mouse 3T6 fibroblasts by 1 to 4 nM soluble alpha-DG. In addition, high-affinity binding to alpha-DG correlated with an ability to infiltrate the white pulp (T-dependent) area of the spleen, cause ablation of the cytotoxic T-lymphocyte (CTL) response by day 7 postinfection, and establish a persistent infection. In contrast, viruses with a lower affinity of binding to alpha-DG were only partially inhibited from infecting alpha-DG(-/-) embryonic stem cells and required a concentration of soluble alpha-DG higher than 100 nM to prevent infection of mouse 3T6 fibroblasts. These viruses that bound at low affinity were mainly restricted to the splenic red pulp, and the host generated an effective CTL response that rapidly cleared the infection. Reassortants of viruses that bound to alpha-DG at high and low affinities were used to map genes responsible for the differences described to the S RNA, containing the virus attachment protein glycoprotein 1.


Subject(s)
Cytoskeletal Proteins/physiology , Lymphocytic choriomeningitis virus/physiology , Membrane Glycoproteins/physiology , Receptors, Virus/physiology , Animals , Dystroglycans , Female , Kinetics , Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred BALB C , RNA, Viral/physiology , Spleen/virology , T-Lymphocytes, Cytotoxic/immunology
15.
J Virol ; 72(11): 8613-9, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9765400

ABSTRACT

The gastric mucosa is an important portal of entry for lymphocytic choriomeningitis virus (LCMV) infections. Within hours after intragastric (i.g.) inoculation, virus appears in the gastric epithelia, then in the mesenteric lymph nodes and spleen, and then in the liver and brain. By 72 h i.g.-inoculated virus is widely disseminated and equivalent to intravenous (i.v.) infection (S. K. Rai, B. K. Micales, M. S. Wu, D. S. Cheung, T. D. Pugh, G. E. Lyons, and M. S. Salvato. Am. J. Pathol. 151:633-639, 1997). Pretreatment of mice with a G protein inhibitor, pertussis toxin (PTx), delays LCMV dissemination after i.g., but not after i.v., inoculation. Delayed infection was confirmed by plaque assays, by reverse transcription-PCR, and by in situ hybridization. The differential PTx effect on i.v. and i.g. infections indicates that dissemination from the gastric mucosa requires signals transduced through heterotrimeric G protein complexes. PTx has no direct effect on LCMV replication, but it modulates integrin expression in part by blocking chemokine signals. LCMV infection of macrophages up-regulates CD11a, and PTx treatment counteracts this. PTx may prevent early LCMV dissemination by inhibiting the G protein-coupled chemotactic response of macrophages infected during the initial exposure, thus blocking systemic virus spread.


Subject(s)
GTP-Binding Proteins/physiology , Gastric Mucosa/virology , Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/pathogenicity , Animals , Cell Adhesion Molecules/metabolism , Cells, Cultured , Chemotaxis/drug effects , DNA Primers/genetics , In Situ Hybridization , Injections, Intravenous , Lymphocytic Choriomeningitis/physiopathology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/physiology , Macrophages/drug effects , Macrophages/physiology , Macrophages/virology , Male , Mice , Mice, Inbred BALB C , Organ Specificity , Pertussis Toxin , Signal Transduction , Time Factors , Virulence Factors, Bordetella/pharmacology , Virus Replication
18.
Virology ; 209(1): 3-9, 1995 May 10.
Article in English | MEDLINE | ID: mdl-7747483

ABSTRACT

The NBD-PE/Rd-PE fluorescent membrane fusion assay was used to measure the pH dependence and kinetics of the fusion activity of lymphocytic choriomeningitis virus with liposomes designed to mimic the composition of the endosomal membrane. Fusion activity was only observed at pH values less than 6.3 and showed a greater rate and extent at lower pH values. Pronounced kinetic fusion curves were observed at pH values below 5.8. When equivalent lipid amounts of target liposomes and virus were mixed at pH 5.3 the dequenching activity had a t1/2 of 45 +/- 10 sec. In addition to catalyzing membrane fusion after acidification the glycoprotein complex was previously found to undergo conformational change (C. Di Simone, M. A. Zandonatti, and M. J. Buchmeier, 1994, Virology 198, 455-465), including loss of the GP-1 polypeptide from the virion surface. The pH dependence and kinetics of this acid-induced GP-1 release were quantitated using centrifugal separation of solubilized GP-1 from pelleted virions. A pH-dependent elution curve was determined with progressively more GP-1 released at pH values below 6.3 and reaching nearly 100% dissociation at pH 5.5 after 30 min at 37 degrees. At pH 5.3 the GP disassembly proceeded with a t1/2 of 7 +/- 2 min. The t1/2 of virus inactivation was also measured at pH 5.3 and 7.0 and found to be 7.9 +/- 1 and 150 min, respectively. Fusion, GP dissociation, and inactivation kinetics data suggest a mechanism in which GP is activated to a fusion active state where membrane lipid exchange occurs and then undergoes an irreversible conformational change which includes the loss of GP-1 from the spike complex.


Subject(s)
Lymphocytic choriomeningitis virus/chemistry , Membrane Glycoproteins/chemistry , Viral Proteins/chemistry , Animals , Humans , Hydrogen-Ion Concentration , Kinetics , Liposomes , Lymphocytic Choriomeningitis/etiology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Membrane Fusion , Molecular Structure
19.
J Neuroimmunol ; 51(2): 147-52, 1994 May.
Article in English | MEDLINE | ID: mdl-8182114

ABSTRACT

The invariably fatal immunopathological disease that follows intracerebral injection of CBA/Ca (H-2k) mice with 1000 PFU of lymphocytic choriomeningitis virus (LCMV) generally fails to develop in congenic mice transgenic for a V beta 8.1D beta 2J beta 2.3C beta 2 T cell receptor (TCR) gene. The majority of these LCMV-infected TCR-transgenic mice show a substantial meningitis of delayed onset, that resolves without causing any obvious clinical impairment. This inflammatory process depends on the involvement of V beta 8+ T cells, but does not require the participation of the CD4+ subset. The cytotoxic effectors that develop in both the transgenic mice and the CBA/Ca controls are lytic for target cells infected with a vaccinia construct expressing genes encoding the putative polymerase protein of LCMV. Limiting the available TCR repertoire to lymphocytes with a single V beta phenotype (not required for the generation of potent effectors in wild-type mice) thus modifies the development of the lethal neuropathology characteristic of LCMV infection, although the CD8+ cytotoxic T lymphocyte response is not greatly compromised.


Subject(s)
Lymphocytic Choriomeningitis/immunology , Receptors, Antigen, T-Cell, alpha-beta/physiology , Acute Disease , Animals , Female , Lymphocytic Choriomeningitis/etiology , Lymphocytic Choriomeningitis/pathology , Male , Mice , Mice, Inbred CBA , Mice, Transgenic , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...