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.
IT has been proposed that lymphocytic choriomeningitis (LCM) virus and immunologically related viruses should be placed in a new taxonomic group, with LCM virus as the prototype virus1,2. They were called arenoviruses because they contain, as a unique feature, electron-dense, sand-like or ribo-some-like granules2. The LCM virus contains RNA and we have previously reported that this RNA can be separated into three components by density gradient centrifugation3.  相似文献   

2.
3.
4.
A quantitative assay for lymphocytic choriomeningitis virus was developed and standardized. The assay is based on direct immunofluorescent staining of infected L-929 cell monolayers and enumeration of cells containing fluorescent viral antigens. Maximal adsorption of virus to cells occurred within 1 h. Observations on the sequential development of viral antigens within cells showed that specific cytoplasmic fluorescence appeared within 10 h. The optimal time for enumerating fluorescent cells was from 18 to 20 h after addition of virus. A linear relationship was demonstrated between the number of infected cells and the relative virus concentration. Fluorescent cells were distributed randomly in infected cover slip cell monolayers. The immunofluorescent cell-counting assay for lymphocytic choriomeningitis virus was highly precise and reproducible.  相似文献   

5.
Pseudotyping can improve retroviral vector stability and transduction efficiency. Here, we describe a novel pseudotype of murine leukemia virus packaged with lymphocytic choriomeningitis virus (LCMV). This pseudotype was stable during ultracentrifugation and infected several cell lines from different species. Moreover, LCMV glycoproteins were not cell toxic.  相似文献   

6.
Lymphocytic choriomeningitis (LCM) virus was observed by electron microscopy in thin sections of infected tissue cultures. The particles were pleomorphic and varied greatly in size. The smaller particles (50 to 200 nm) appeared to be spherical, whereas the largest (over 200 nm) were often cup-shaped. All particles contained one to eight or more electron-dense granules which were removed by ribonuclease. The particles were formed by budding from the plasma membrane and appeared to have spikes. The morphological evidence suggests that LCM should be considered as belonging to the presently unclassified group of lipoprotein-enveloped ribonucleic acid viruses.  相似文献   

7.
The viral family Arenaviridae includes a number of viruses that can cause hemorrhagic fever in humans. Arenavirus infection often involves multiple organs and can lead to capillary instability, impaired hemostasis, and death. Preclinical testing for development of antiviral or therapeutics is in part hampered due to a lack of an immunologically well-defined rodent model that exhibits similar acute hemorrhagic illness or sequelae compared to the human disease. We have identified the FVB mouse strain, which succumbs to a hemorrhagic fever-like illness when infected with lymphocytic choriomeningitis virus (LCMV). FVB mice infected with LCMV demonstrate high mortality associated with thrombocytopenia, hepatocellular and splenic necrosis, and cutaneous hemorrhage. Investigation of inflammatory mediators revealed increased IFN-γ, IL-6 and IL-17, along with increased chemokine production, at early times after LCMV infection, which suggests that a viral-induced host immune response is the cause of the pathology. Depletion of T cells at time of infection prevented mortality in all treated animals. Antisense-targeted reduction of IL-17 cytokine responsiveness provided significant protection from hemorrhagic pathology. F1 mice derived from FVB×C57BL/6 mating exhibit disease signs and mortality concomitant with the FVB challenged mice, extending this model to more widely available immunological tools. This report offers a novel animal model for arenavirus research and pre-clinical therapeutic testing.  相似文献   

8.
Following infection of mice with lymphocytic choriomeningitis virus (LCMV), virus-neutralizing antibodies appear late, after 30 to 60 days. Such neutralizing antibodies play an important role in protection against reinfection. To analyze whether a neutralizing antibody response which developed earlier could contribute to LCMV clearance during the acute phase of infection, we generated transgenic mice expressing LCMV-neutralizing antibodies. Transgenic mice expressing the immunoglobulin μ heavy chain of the LCMV-neutralizing monoclonal antibody KL25 (H25 transgenic mice) mounted LCMV-neutralizing immunoglobulin M (IgM) serum titers within 8 days after infection. This early inducible LCMV-neutralizing antibody response significantly improved the host’s capacity to clear the infection and did not cause an enhancement of disease after intracerebral (i.c.) LCMV infection. In contrast, mice which had been passively administered LCMV-neutralizing antibodies and transgenic mice exhibiting spontaneous LCMV-neutralizing IgM serum titers (HL25 transgenic mice expressing the immunoglobulin μ heavy and the κ light chain) showed an enhancement of disease after i.c. LCMV infection. Thus, early-inducible LCMV-neutralizing antibodies can contribute to viral clearance in the acute phase of the infection and do not cause antibody-dependent enhancement of disease.Against many cytopathic viruses such as poliovirus, influenza virus, rabies virus, and vesicular stomatitis virus, protective virus-neutralizing antibodies are generated early, within 1 week after infection (3, 31, 36, 44, 49). In contrast, several noncytopathic viruses (e.g., human immunodeficiency virus and hepatitis viruses B and C in humans or lymphocytic choriomeningitis virus [LCMV] in mice) elicit poor and delayed virus-neutralizing antibody responses (1, 7, 20, 24, 27, 35, 45, 48).In the mouse, the natural host of LCMV, the acute LCMV infection is predominantly controlled by cytotoxic T lymphocytes (CTLs) in an obligatory perforin-dependent manner (13, 18, 28, 50). In addition to the CTL response, LCMV-specific antibodies are generated. Early after infection (by day 8), a strong antibody response specific for the internal viral nucleoprotein (NP) is mounted (7, 19, 23, 28). These early LCMV NP-specific antibodies exhibit no virus-neutralizing capacity (7, 10). Results from studies of B-cell-depleted mice and B-cell-deficient mice implied that the early LCMV NP-specific antibodies are not involved in the clearance of LCMV (8, 11, 12, 40). Late after infection (between days 30 and day 60), LCMV-neutralizing antibodies develop (7, 19, 22, 28, 33); these antibodies are directed against the surface glycoprotein (GP) of LCMV (9, 10). LCMV-neutralizing antibodies have an important function in protection against reinfection (4, 6, 38, 41, 47).In some viral infections, subprotective virus-neutralizing antibody titers can enhance disease rather than promote host recovery (i.e., exhibit antibody-dependent enhancement of disease [ADE] [14, 15, 21, 46]). For example, neutralizing antibodies are involved in the resolution of a primary dengue virus infection and in the protection against reinfection. However, if subprotective neutralizing antibody titers are present at the time of reinfection, a severe form of the disease (dengue hemorrhagic fever/dengue shock syndrome [15, 21]), which might be caused by Fc receptor-mediated uptake of virus-antibody complexes leading to an enhanced infection of monocytes (15, 16, 25, 39), can develop. Similarly, an enhancement of disease after intracerebral (i.c.) LCMV infection was observed in mice which had been treated with virus-neutralizing antibodies before the virus challenge (6). ADE in LCMV-infected mice was either due to an enhanced infection of monocytes by Fc receptor-mediated uptake of antibody-virus complexes or due to CTL-mediated immunopathology caused by an imbalanced virus spread and CTL response.To analyze whether LCMV-neutralizing antibodies generated early after infection improve the host’s capacity to clear the virus or enhance immunopathological disease, immunoglobulin (Ig)-transgenic mice expressing LCMV-neutralizing IgM antibodies were generated. After LCMV infection of transgenic mice expressing the Ig heavy chain (H25 transgenic mice), LCMV-neutralizing serum antibodies were mounted within 8 days, which significantly improved the host’s capacity to eliminate LCMV. H25 transgenic mice did not show any signs of ADE after i.c. LCMV infection.Transgenic mice expressing the Ig heavy and light chains (HL25 transgenic mice) exhibited spontaneous LCMV-neutralizing serum antibodies and confirmed the protective role of preexisting LCMV-neutralizing antibodies, even though the neutralizing serum antibodies were of the IgM isotype. Similar to mice which had been treated with LCMV-neutralizing antibodies, HL25 transgenic mice developed an enhanced disease after i.c. LCMV infection, which indicated that ADE was due to an imbalance between virus spread and CTL response. Thus, the early-inducible LCMV-neutralizing antibody response significantly enhanced clearance of the acute infection without any risk of causing ADE.  相似文献   

9.
目的用重组蛋白抗原取代淋巴细胞脉络丛脑膜炎病毒(LCMV)抗原用于实验动物感染LCMV检测.方法合成LCMVsRNA第1816~2178位核苷酸序列编码的Np第380~500位氨基酸基因,克隆在Hisx6-GSTpET-28a载体中,表达产物经固定化金属配体亲和层析纯化,建立ELISA检测试剂.结果合成基因表达产物经Ni-NTA-Agarose纯化后纯度达到95%以上.ELISA检测小鼠、地鼠和豚鼠结果与全病毒抗原基本一致,而且特异性较强,操作简便.结论用合成基因表达产物取代LCMV抗原检测病毒抗体,不仅可以消除病毒传播可能,而且特异性强.为实验动物及生物材料质量控制提供安全有效方法.  相似文献   

10.
Purified preparations of lymphocytic choriomeningitis virus (LCM virus) contain three classes of RNA. The previously described 18s, 23s, 28s, and 31s RNAs, where the 23s and 31s RNAs are viral-specific, and the 18s and 28s RNAs probably are host RNAs incorporated in the virion. Now, 4s, 5s, and 5.5s RNAs can be isolated as well. Thus five RNAs which migrate by acrylamide gel electrophoresis as ribosome-derived RNA can be isolated from purified LCM virus. This observation further supports the reports that arenaviruses may contain ribosomes. The ribosome-derived RNA can be synthesized both before and after the virus infection. The viral 23s could be a hydrogen-bonded complex forming the 31s RNA, or it could be contained in defective interfering LCM virus particles; these possibilities are examined.  相似文献   

11.
All of the ten strains of lymphocytic choriomeningitis virus assayed on BHK 21/13S cells showed various degrees of plaque size heterogeneity. The amount of virus released from these plaques was usually very small because of rapid photodynamic inactivation by neutral red. When virus from large and small plaques of a specific strain was plated, the same distribution of plaque size was obtained from each clone. Although it was shown that surface virus could possibly be randomly distributed at the time of addition of neutral red overlays, no virus could be isolated from nonplaque areas. Two different strains of virus (CA1371 and WE) with markedly different plaque size ranges were separated by plaque excision from plates infected with a mixture of both viruses.  相似文献   

12.
13.
By means of the indirect fluorescent-antibody test, cross serological reactivity was demonstrated between lymphocytic choriomeningitis (LCM) virus and the viruses of the Tacaribe complex. Antisera to all members of the Tacaribe complex reacted with LCM virus; LCM antisera gave significant staining of Amapari virus, but minimal or inconsistent reactions with Tacaribe virus, and no reaction with two other viruses of the Tacaribe complex. A low level cross-reaction was observed in complement fixation tests of Machupo and Pichinde antisera against LCM antigen. Immunization with Tacaribe and Amapari viruses did not protect mice against challenge with LCM virus. Because of the identical appearance of the virions, the sharing of antigens, and the many biological similarities between LCM and the Tacaribe complex viruses, it is proposed that they be considered as constituting a new taxonomic group of viruses.  相似文献   

14.
Infection of BHK-21 cells with lymphocytic choriomeningitis (LCM) virus resulted in the production of significant titers of complement-fixing (CF) antigen. The antigen was spontaneously released from the cells, but the highest titer of 1:16 was recovered by disruption of the infected cells by freeze-thawing in tryptose phosphate broth. The antigen could be partially separated from infectious virus by centrifugation. Furthermore, it was possible to detect LCM virus infection of cell cultures by the production of the CF antigen, but this method proved less sensitive than titration by intracerebral inoculation of mice. The CF antigen from cell cultures was at least as sensitive and specific as the reference antigen prepared from infected guinea pig spleen.  相似文献   

15.
Type I interferons (IFNs) play a critical role in the host defense against viruses. Lymphocytic choriomeningitis virus (LCMV) infection induces robust type I IFN production in its natural host, the mouse. However, the mechanisms underlying the induction of type I IFNs in response to LCMV infection have not yet been clearly defined. In the present study, we demonstrate that IRF7 is required for both the early phase (day 1 postinfection) and the late phase (day 2 postinfection) of the type I IFN response to LCMV, and melanoma differentiation-associated gene 5 (MDA5)/mitochondrial antiviral signaling protein (MAVS) signaling is crucial for the late phase of the type I IFN response to LCMV. We further demonstrate that LCMV genomic RNA itself (without other LCMV components) is able to induce type I IFN responses in various cell types by activation of the RNA helicases retinoic acid-inducible gene I (RIG-I) and MDA5. We also show that expression of the LCMV nucleoprotein (NP) inhibits the type I IFN response induced by LCMV RNA and other RIG-I/MDA5 ligands. These virus-host interactions may play important roles in the pathogeneses of LCMV and other human arenavirus diseases.Type I interferons (IFNs), namely, alpha interferon (IFN-α) and IFN-β, are not only essential for host innate defense against viral pathogens but also critically modulate the development of virus-specific adaptive immune responses (6, 8, 28, 30, 36, 50, 61). The importance of type I IFNs in host defense has been demonstrated by studying mice deficient in the type I IFN receptor, which are highly susceptible to most viral pathogens (2, 47, 62).Recent studies have suggested that the production of type I IFNs is controlled by different innate pattern recognition receptors (PRRs) (19, 32, 55, 60). There are three major classes of PRRs, including Toll-like receptors (TLRs) (3, 40), retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) (25, 48, 51), and nucleotide oligomerization domain (NOD)-like receptors (9, 22). TLRs are a group of transmembrane proteins expressed on either cell surfaces or endosomal compartments. RLRs localize in the cytosol. Both TLRs and RLRs are involved in detecting viral pathogens and controlling the production of type I IFNs (52, 60). In particular, the endosome-localized TLRs (TLR3, TLR7/8, and TLR9) play important roles in detecting virus-derived double-stranded RNA (dsRNA), single-stranded RNA (ssRNA), and DNA-containing unmethylated CpG motifs, respectively. In contrast, RIG-I detects virus-derived ssRNA with 5′-triphosphates (5′-PPPs) or short dsRNA (<1 kb), whereas melanoma differentiation-associated gene 5 (MDA5) is responsible for recognizing virus-derived long dsRNA as well as a synthetic mimic of viral dsRNA poly(I):poly(C) [poly(I·C)] (24, 60). Recognition of viral pathogen-associated molecular patterns (PAMPs) ultimately leads to the activation and nuclear translocation of interferon regulatory factors (IRFs) and nuclear factor κB (NF-κB), which, in turn, switches on a cascade of genes controlling the production of both type I IFNs and other proinflammatory cytokines (10, 11, 60).Lymphocytic choriomeningitis virus (LCMV) infection in its natural host, the mouse, is an excellent system to study the impact of virus-host interactions on viral pathogenesis and to address important issues related to human viral diseases (1, 45, 49, 67). LCMV infection induces type I IFNs as well as other proinflammatory chemokines and cytokines (6, 41). Our previous studies have demonstrated that TLR2, TLR6, and CD14 are involved in LCMV-induced proinflammatory chemokines and cytokines (66). The mechanism by which LCMV induces type I IFN responses, however, has not been clearly defined (7, 8, 31, 44). The role of the helicase family members RIG-I and MDA5 in virus-induced type I IFN responses has been recently established. RIG-I has been found to be critical in controlling the production of type I IFN in response to a number of RNA viruses, including influenza virus, rabies virus, Hantaan virus, vesicular stomatitis virus (VSV), Sendai virus (SeV), etc. In contrast, MDA5 is required for responses to picornaviruses (15, 25, 63).In the present study, we demonstrated that LCMV genomic RNA strongly activates type I IFNs through a RIG-I/MDA5-dependent signaling pathway. Our present study further demonstrated that the LCMV nucleoprotein (NP) blocks LCMV RNA- and other viral ligand-induced type I IFN responses.  相似文献   

16.
Several arenavirus pathogens, such as Lassa and Junin viruses, inhibit macrophage activation, the molecular mechanism of which is unclear. We show that lymphocytic choriomeningitis virus (LCMV) can also inhibit macrophage activation, in contrast to Pichinde and Tacaribe viruses, which are not known to naturally cause human diseases. Using a recombinant Pichinde virus system, we show that the LCMV Z N-terminal domain (NTD) mediates the inhibition of macrophage activation and immune functions.  相似文献   

17.
Biological, biochemical, and biophysical properties of three lymphocytic choriomeningitis (LCM) virus strains were compared. The biological property examined was the concentration range of virus which would, when injected into neonates, cause a carrier state. The dosage range for the CA1371 and Traub strains was found to be as broad as the limits examined (5 to 100 ld(50) units/mouse). The WCP strain, however, would only produce carriers within a 3 to 5 ld(50) range. The biochemical properties examined were the growth rates in tissue culture and the effect of varying the input ratio of virus to cells. With identical input ratios, the Traub strain reached a peak titer 32 hr after infection. The CA1371 and WCP strain reached their peaks at the 40th hr. With a 10-fold decrease in the amount of CA1371 virus per cell, peak titer (as high as in the above experiments) was not obtained until 56 hr postinfection. The biophysical properties examined were stability in density gradients and inactivation rates at 4C. In potassium tartrate gradients, full recovery of the CA1371 and WCP strain could be achieved. However, inactivation kinetics showed that only the CA1371 strain was much more stable than the Traub-LCM. The realization that marked differences in LCM strains exist is discussed in relation to certain inconsistencies in the literature.  相似文献   

18.
19.
Lymphocytic choriomeningitis (LCM) virus, Traub strain, was purified from BHK-21 tissue culture medium. The virus was then analyzed by equilibrium centrifugation and rate zonal centrifugation in sucrose gradients. A buoyant density in sucrose of 1.18 g/ml was found and the S(20, w) value was estimated to be about 470 to 500S. Furthermore, the (3)H-uridine-labeled ribonucleic acid (RNA) from virus was extracted from LCM virus and analyzed by rate zonal centrifugation. Two major and one minor single-stranded RNA components were found with sedimentation coefficients of 28, 22, and 18S.  相似文献   

20.
The yields of the Armstrong and WE strains of lymphocytic choriomeningitis virus in baby hamster kidney (BHK) cells cultivated in either bovine, calf, fetal bovine, or horse serum were investigated. Lines of BHK cells were established in these sera. When the infected cell lines were observed by immunofluorescence, the per cent fluorescing cells for a given virus strain did not vary. However, for both strains, the extracellular virus yields per cell were significantly greater in the fetal bovine-cell line than in the other serum-cell lines.  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