首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
2.
Rojek JM  Perez M  Kunz S 《Journal of virology》2008,82(3):1505-1517
In contrast to most enveloped viruses that enter the host cell via clathrin-dependent endocytosis, the Old World arenavirus lymphocytic choriomeningitis virus (LCMV) enters cells via noncoated vesicles that deliver the virus to endosomes, where pH-dependent membrane fusion occurs. Here, we investigated the initial steps of LCMV infection. We found that the attachment of LCMV to its cellular receptor α-dystroglycan occurs rapidly and is not dependent on membrane cholesterol. However, subsequent virus internalization is sensitive to cholesterol depletion, indicating the involvement of a cholesterol-dependent pathway. We provide evidence that LCMV entry involves an endocytotic pathway that is independent of clathrin and caveolin and that does not require the GTPase dynamin. In addition, neither the structural integrity nor the dynamics of the actin cytoskeleton are required for infection. These findings indicate that the prototypic Old World arenavirus LCMV uses a mechanism of entry that is different from clathrin-mediated endocytosis, which is used by the New World arenavirus Junin virus, and pathways used by other enveloped viruses.  相似文献   

3.
Lymphocytic choriomeningitis virus (LCMV) is a zoonotic pathogen of which mice are the natural reservoir. Different strains and clones of LCMV show different pathogenicity in mice. Here we determined the complete genomic sequences of 3 LCMV strains (OQ28 and BRC which were isolated from mice in Japan and WE(ngs) which was derived from strain WE). Strains OQ28 and BRC showed high sequence homology with other LCMV strains. Although phylogenetic analyses placed these 2 Japanese strains in different subclusters, they belonged to same cluster of LCMV isolates. WE(ngs) and WE had many sequence substitutions between them but fell into same subcluster. The pathogenicity of the 3 new LCMV isolates was examined by inoculating ICR mice with 102 and 104 TCID50 of virus. ICR mice infected with OQ28 or WE(ngs) exhibited severe clinical signs, and some of the infected mice died. In contrast, all ICR mice infected with BRC showed no clinical signs and survived infection. Virus was detected in the blood, organs, or both of most of the surviving ICR mice inoculated with either OQ28 or WE(ngs). However, virus was below the level of detection in all ICR mice surviving infection with strain BRC. Therefore, LCMV strains OQ28 and BRC were genetically classified in the same cluster of LCMV strains but exhibited very different pathogenicity.Abbreviations: dpi, days postinfection; GP, viral glycoprotein; h, hydrophobic region; IFA, indirect fluorescent antibody assay; L, viral RNA-dependent RNA polymerase; LCMV, lymphocytic choriomeningitis virus; NP, nucleocapsid protein; UTR, untranslated region; Z, zinc-finger proteinLymphocytic choriomeningitis virus (LCMV) is a member of the genus Arenavirus in the family Arenaviridae. The genus Arenavirus is divided into 2 groups (Old World and New World arenaviruses) according to genetic and antigenic characteristics.4 LCMV is a member of the Old World arenavirus group, which also includes Lassa, Mopeia, Mobala, and Ippy viruses.4,10 The LCMV genome contains 2 negative-sense single-stranded RNA segments, designated S RNA and L RNA, with approximate sizes of 3.4 kb and 7.2 kb, respectively.30,31 Each RNA segment has an ambisense coding strategy, encoding 2 different proteins in opposite orientations. S RNA encodes the nucleocapsid protein and glycoprotein, and L RNA encodes the viral RNA-dependent RNA polymerase and a small zinc finger protein.25,30LCMV is a zoonotic agent that is transmitted to humans via urine or saliva of infected mice (Mus musculus), which are a natural reservoir of the virus.4 The prevalence of LCMV in mice is 7.0% to 25.9% in Japan and 4% to 9% in Europe.5,17,19,20,35 Mice are naturally infected by either vertical or horizontal transmission of the virus, and infected mice usually show no clinical signs. In contrast, experimentally infected mice inoculated intraperitoneally or intracerebrally can exhibit clinical signs such as ruffled fur, half-closed eyes, hunched posture, immobility, and neurologic deficits.4,12,19 Although human LCMV infections are generally either asymptomatic or mild, immunodeficient persons can develop spontaneous abortion, severe birth defects, aseptic meningitis, or fatal infections.1,2,13,22,27 Therefore, LCMV is an important agent that should be monitored in facilities housing and breeding mice.LCMV strains Armstrong, Traub, and WE were isolated during the 1930s from laboratory mice and humans working in a mouse facility.4 Many other LCMV strains and clones used in research originated from these 3 isolates. Strains Aggressive and Docile are clones (variants) of strain UBC, which was derived from the parental strain WE, and strains E350, CA1371, 53b, and clone 13 were all derived from strain Armstrong.4 The lethality of strains Aggressive and Docile varies between mouse strains.38 Mice inoculated with 53b develop acute infections, whereas those inoculated with clone 13 mount chronic infections, even though both of the strains were derived from strain Armstrong.29 Furthermore, strain Armstrong produces more severe disease in C3H mice than do strains WE and Traub.4 Therefore, previous studies indicate that mice infected with different strains of LCMV exhibit differences in clinical signs and lethality.4,7 LCMV is a noncytolytic virus and causes immune-mediated viral disease.12 The clinical signs and lethal disease arise because virus-specific T cells attack infected cells on critical organs in infected mice.12Here we report the characterization of 2 LCMV strains recently isolated in Japan (strains OQ28 and BRC) and a passaged isolate of strain WE. The complete genomic sequences of these 3 strains were determined, and their phylogenetic relationship to other LCMV strains was assessed. We also evaluated the pathogenicity in ICR mice of these isolates.  相似文献   

4.
5.
6.
The role of antibody in immune recovery from infection with lymphocytic choriomeningitis virus (LCMV) strain WE was evaluated in B-cell-depleted mice. Mice were treated from birth with either affinity-purified rabbit anti-mouse immunoglobulin M (IgM), normal rabbit immunoglobulin, or, alternatively, an affinity-purified monoclonal rat anti-mouse IgM antibody (LO-MM-9); untreated mice served as controls. B-cell depletion was considered complete in specifically treated mice according to the following criteria: absence of a significant response to the B-cell mitogen lipopolysaccharide, absence of B cells expressing immunoglobulin on their surfaces, absence of detectable IgM or IgG in serum, and presence in the serum of free anti-IgM antibodies. In organs of mu-suppressed BALB/c mice, LCMV-WE replicated, dependent upon organ, at the same rate or more rapidly and, in general, to higher titers than in normal rabbit immunoglobulin-treated mice; untreated mice eliminated the virus most rapidly and showed lower virus titers. In addition, LCMV-primed control mice cleared a second LCMV challenge very rapidly and contained no virus by day 3, whereas mu-suppressed mice had virus in their blood and organs (except the spleen) up to days 3 to 6. The observed effects of anti-mu treatment may reflect the action of neutralizing antibodies (which so far have been difficult to demonstrate in vivo) or other antibody-dependent antiviral mechanisms which, together with T cells, efficiently control LCMV clearance.  相似文献   

7.
Laboratory studies of arenaviruses have been limited to parenteral routes of infection; however, recent epidemiological studies implicate virus ingestion as a natural route of infection. Accordingly, we developed a model for oral and gastric infection with lymphocytic choriomeningitis virus to enable studies of mucosal transmission and vaccination by this additional route.  相似文献   

8.
Infection with virus variants exhibiting changes in the peptide sequences defining immunodominant determinants that abolish recognition by antiviral cytotoxic T cells (CTL) presents a considerable challenge to the antiviral T-cell immune system and may enable some viruses to persist in hosts. The potential importance of such variants with respect to mechanisms of viral persistence and disease pathogenesis was assessed by infecting adult mice with variants of lymphocytic choriomeningitis virus (LCMV) strain WE. These variants were selected in vivo or in vitro for resistance to lysis by CD8+ H-2b-restricted antiviral CTL. The majority of anti-LCMV CTL in infected H-2b mice recognize epitopes defined by residues 32 to 42 and 275 to 289 (epitopes 32-42 and 275-289) of the LCMV glycoprotein or 397 to 407 of the viral nucleoprotein. The 8.7 variant exhibits a change in the epitope 32-42 (Val-35-->Leu), and variant CL1.2 exhibits a change in the epitope 275-289 (Asn-280-->Asp) of the wild-type LCMV-WE. The double-mutated 8.7-B23 variant had the variation of 8.7 and an additional change located in the epitope 275-289 (Asn-280-->Ser). The 8.7 variant peptide with unchanged anchor positions bound efficiently to H-2Db and H-2Kb molecules but induced only a very weak CTL response. CTL epitope 275-289 of CL1.2 and 8.7-B23 altered at predicted anchor residues were unable to bind Db molecules and were also not recognized by antiviral CTL. Infection of C57BL/6 mice (H-2b) with the variants exhibiting mutations of one of the CTL epitopes, i.e., 8.7 or CL1.2, induced CTL responses specific for the unmutated epitopes comparable with those induced by infection with WE, and these responses were sufficient to eliminate virus from the host. In contrast, infection with the double-mutated variant 8.7-B23 induced CTL activity that was reduced by a factor of about 50-fold compared with wild-type LCMV. Consequently, high doses (10(7) PFU intravenously) of this virus were eliminated slowly and only by about day 100 after infection. 8.7-B23 failed to cause lethal lymphocytic choriomeningitis after intracerebral infection with a dose of > 10(4) PFU in C57BL/6 mice (but not in mice of nonselecting H-2d haplotype); with the other variants or wild-type LCMV, doses greater than 10(6) to 10(7) PFU were necessary to avoid lethal choriomeningitis.(ABSTRACT TRUNCATED AT 400 WORDS)  相似文献   

9.
Mice 2--4 days of age were pretreated with a single 5 mg/kg dose of dianhydrodulcitol (DAD) and later infected intracerebrally with lymphocytic choriomeningitis (LCM) virus. These animals had a lower mortality rate and died later than the untreated control animals. Thus DAD pretreatment prevented in part of the animals the development of lethal meningitis, the consequence of LCM virus infection, reducing the cellular immune response. This effect of DAD could equally be observed in animals infected at the age of 16--18 days and of 4 weeks.  相似文献   

10.
11.
The parameters of cell-mediated immune responses of rats to infection with lymphocytic choriomeningitis virus or vaccinia virus were assessed by measuring primary footpad swelling, increased weights of the local lymph nodes, increased numbers of lymphocytes per lymph node, and the course of virus-specific cytolytic activity by these lymphocytes. Except for lack of a defined swelling caused by vaccinia virus injected into the hind footpads of rats, the kinetics of all these responses correlated and were in accord with the usual time course of cellular immune responses. Starting 3 days after infection, peaking at 5 to 7 days, and disappearing after 10 to 12 days, the responses by rats to both viruses were comparable to those found in mice. The phagocytes of these infected rats inhibited the growth of Listeria monocytogenes in vivo, indicating activation of the macrophages by virus-specific cellular immunity. The rat cytotoxic lymphocytes were thymus derived as judged by various criteria: inactivation by an absorbed rabbit anti-rat brain antiserum plus C, susceptibility to anti Thy 1.1 plus C, restriction of the lytic activity within inbred strains and probably by the Ag-B locus, and the kinetics of the response. The cytotoxic T lymphocytes were virus specific since they killed only target cells infected with the same virus but not uninfected cells, or targets that were infected with an unrelated virus.  相似文献   

12.
13.
14.
Signal peptides (SPs) direct nascent secretory and membrane proteins to the membrane of the endoplasmic reticulum. They are usually cleaved from the nascent polypeptide by signal peptidase and then further proteolytically processed. The SP of the pre-glycoprotein (pGP-C) of the lymphocytic choriomeningitis virus SPGP-C (signal peptide of pGP-C) shows different properties: 1) The SPGP-C is unusually long (58 amino acid residues) and contains two hydrophobic segments interrupted by a lysine residue. 2) The SPGP-C is cleaved only from a subset of pGP-C proteins. A substantial portion of pGP-C accumulates that still contains the SPGP-C.3)The cleaved SPGP-C is rather long-lived (t(1/2) of more than 6 h). 4) The cleaved SPGP-C resides in the membrane and is resistant to digestion with proteinase K even in the presence of detergents, suggesting a very compact structure. 5) SPGP-C accumulates in virus particles. These unusual features of the cleaved SPGP-C suggest that SPGP-C not only targets the nascent pGP-C to the endoplasmic reticulum membrane but also has additional functions in lymphocytic choriomeningitis virus life cycle.  相似文献   

15.
DNA vaccination has been evaluated with the lymphocytic choriomeningitis virus (LCMV) model system. Plasmid DNA encoding the LCMV nucleoprotein, when injected intramuscularly, induces both antiviral antibodies and cytotoxic T lymphocytes. Injection of DNA encoding the nucleoprotein or the viral glycoprotein confers protection against normally lethal LCMV challenge in a major histocompatibility complex-dependent manner. The protection conferred is incomplete, but it is most probably mediated by the induced cytotoxic T lymphocytes.  相似文献   

16.
The role of gamma interferon (IFN-gamma) induced during a viral infection in the ability of the host to acquire antiviral immunity was studied in mice. They were injected subcutaneously daily with an ammonium sulfate-precipitated sheep anti-IFN-gamma antibody preparation able to neutralize 10(4) U of IFN-gamma. Specificity of the anti-IFN-gamma antiserum was demonstrated by absence of detectable activity against natural IFN-alpha and -beta. Controls were treated with a similarly prepared normal sheep serum. Treatment with the IFN-gamma-specific antibody preparation had no influence on the ability of mice to generate anti-vaccinia virus- or anti-vesicular stomatitis virus (VSV)-specific cytotoxic T-cell (CTL) responses or T helper-dependent immunoglobulin G responses to VSV. In contrast, treatment of mice with sheep anti-IFN-gamma impaired CTL responses against lymphocytic choriomeningitis (LCM) virus (LCMV, Aggressive isolate); in addition, under the experimental conditions used, it prevented lethal LCM. Cytotoxic T-cell activity measured in the spleens of anti-IFN-gamma-treated mice was comparable to that found in mice initially infected with a 100-fold-larger dose of LCMV. Evaluation of the effects of treatment on the kinetics of virus replication revealed that in both euthymic and athymic nude C57BL/6 mice, anti-IFN-gamma treatment led to an increase of virus titers up to 100-fold compared with control mice. Therefore, IFN-gamma may play a role in controlling viruses with tropism for lymphocytes and monocytes/macrophages, such as LCMV.  相似文献   

17.
The interferon-inducible adenosine deaminase that acts on double-stranded RNA (ADAR1-L) has been proposed to be one of the antiviral effector proteins within the complex innate immune response. Here, the potential role of ADAR1-L in the innate immune response to lymphocytic choriomeningitis virus (LCMV), a widely used virus model, was studied. Infection with LCMV clearly upregulated ADAR1-L expression and activity. The editing activity of ADAR1-L on an RNA substrate was not inhibited by LCMV replication. Accordingly, an adenosine-to-guanosine (A-to-G) and uracil-to-cytidine (U-to-C) hypermutation pattern was found in the LCMV genomic RNA in infected cell lines and in mice. In addition, two hypermutated clones with a high level of A-to-G or U-to-C mutations within a short stretch of the viral genome were isolated. Analysis of the functionality of viral glycoprotein revealed that A-to-G- and U-to-C-mutated LCMV genomes coded for nonfunctional glycoprotein at a surprisingly high frequency. Approximately half the GP clones with an amino acid mutation lacked functionality. These results suggest that ADAR1-L-induced mutations in the viral RNA lead to a loss of viral protein function and reduced viral infectivity. This study therefore provides strong support for the contribution of ADAR1-L to the innate antiviral immune response.  相似文献   

18.
19.
Cluster of differentiation (CD)8+ T cells are like a double edged sword during chronic viral infections because they not only promote virus elimination but also induce virus-mediated immunopathology. Elevated levels of reactive oxygen species (ROS) have been reported during virus infections. However, the role of ROS in T-cell-mediated immunopathology remains unclear. Here we used the murine lymphocytic choriomeningitis virus to explore the role of ROS during the processes of virus elimination and induction of immunopathology. We found that virus infection led to elevated levels of ROS producing granulocytes and macrophages in virus-infected liver and spleen tissues that were triggered by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Lack of the regulatory subunit p47phox of the NADPH oxidase diminished ROS production in these cells. While CD8+ T cells exhibited ROS production that was independent of NADPH oxidase expression, survival and T-cell function was elevated in p47phox-deficient (Ncf1−/−) mice. In the absence of p47phox, enhanced T-cell immunity promoted virus elimination and blunted corresponding immunopathology. In conclusion, we find that NADPH-mediated production of ROS critically impairs the immune response, impacting elimination of virus and outcome of liver cell damage.  相似文献   

20.
The course of intracerebral lymphocytic choriomeningitis (LCM) virus infection was studied in mice treated simultaneously with dianhydrodulcitol (DAD) and phytohaemagglutinin (PHA). Earlier experiments revealed that DAD decreased and PHA enhanced the cellular immune response of mice to LCM virus infection. On applying the treatments simultaneously they inhibited each other and neither the decreasing effect of DAD nor the enhancing effect of PHA on the cellular immune response could be observed.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号