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 990 毫秒
1.
2.
3.
4.
5.
6.
7.
Lassa virus is an enveloped virus with glycoprotein spikes on its surface. It contains an RNA ambisense genome that encodes the glycoprotein precursor GP-C, the nucleoprotein NP, the polymerase L, and the Z protein. Here we demonstrate that the Lassa virus Z protein (i). is abundant in viral particles, (ii). is strongly membrane associated, (iii). is sufficient in the absence of all other viral proteins to release enveloped particles, and (iv). contains two late domains, PTAP and PPXY, necessary for the release of virus-like particles. Our data provide evidence that Z is the Lassa virus matrix protein that is the driving force for virus particle release.  相似文献   

8.
Several arenaviruses, including Lassa fever virus, cause severe, often lethal hemorrhagic fever in humans. No licensed vaccines are available in the United States, and currently there is no efficacious therapy to treat this viral infection. Therefore the importance of developing effective antiviral approaches to combat pathogenic arenaviruses is clear. Moreover, the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is an important model for the study of viral persistence and associated diseases, as well as for exploring therapies to treat viral chronic infections. The use of small interfering RNAs (siRNAs) to downregulate gene expression via RNA interference (RNAi) has emerged as a powerful genetic tool for the study of gene function. In addition, the successful use of siRNAs to target a variety of animal viruses has led us to consider RNAi as a potential novel antiviral strategy. We have investigated the use of RNAi therapy against LCMV. Here, we show that siRNAs targeting sequences within the viral L polymerase and Z mRNAs inhibit LCMV multiplication in cultured cells. Unexpectedly, the antiviral efficacy of RNAi-based therapy against LCMV was highly dependent on the method used to deliver effector siRNA molecules. Thus, transfection of chemically synthesized siRNA pools to L and Z was ineffective in preventing virus multiplication. In contrast, targeting of the same viral L and Z gene products with siRNAs produced inside cells using a replication-deficient recombinant adenovirus expression system inhibited LCMV multiplication very efficiently. Notably, transduction with the replication-deficient recombinant adenovirus expression system to Z and L effectively cured persistently LCMV-infected cells, suggesting the feasibility of using RNAi therapy to combat viral chronic infections by riboviruses.  相似文献   

9.
We have used a reverse genetic approach to identify the viral proteins required for packaging and assembly of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). Plasmids encoding individual LCMV proteins under the control of an RNA polymerase II promoter were cotransfected with a plasmid containing an LCMV minigenome (MG). Intracellular synthesis of the LCMV MG was driven by T7 RNA polymerase whose expression was also mediated by a Pol II promoter. The supernatant from transfected cells was passaged onto fresh cells that were subsequently infected with LCMV to provide the minimal viral trans-acting factors, NP and L, that are required for LCMV MG RNA replication and expression. Reconstitution of LCMV-specific packaging and passage was detected by expression of the chloramphenicol acetyl transferase (CAT) reporter gene present in the MG. NP and L did not direct detectable levels of MG passage. Addition of Z and GP resulted in high levels of passage of CAT activity, which could be prevented by LCMV neutralizing antibodies. Passage of LCMV MG was inhibited by omission of either GP or Z.  相似文献   

10.
Ebola virus (EBOV) is a key member of Filoviridae family and causes severe human infectious diseases with high morbidity and mortality. As a typical negative-sense single-stranded RNA (-ssRNA) viruses, EBOV possess a nucleocapsid protein (NP) to facilitate genomic RNA encapsidation to form viral ribonucleoprotein complex (RNP) together with genome RNA and polymerase, which plays the most essential role in virus proliferation cycle. However, the mechanism of EBOV RNP formation remains unclear. In this work, we solved the high resolution structure of core domain of EBOV NP. The polypeptide of EBOV NP core domain (NPcore) possesses an N-lobe and C-lobe to clamp a RNA binding groove, presenting similarities with the structures of the other reported viral NPs encoded by the members from Mononegavirales order. Most strikingly, a hydrophobic pocket at the surface of the C-lobe is occupied by an α-helix of EBOV NPcore itself, which is highly conserved among filoviridae family. Combined with other biochemical and biophysical evidences, our results provides great potential for understanding the mechanism underlying EBOV RNP formation via the mobility of EBOV NP element and enables the development of antiviral therapies targeting EBOV RNP formation.  相似文献   

11.
12.
Several arenaviruses, chiefly Lassa virus (LASV) and Junin virus in West Africa and Argentina, respectively, cause hemorrhagic fever (HF) disease in humans that is associated with high morbidity and significant mortality. The investigation of antiviral strategies to combat HF arenaviruses is hampered by the requirement of biosafety level 4 (BSL-4) facilities to work with these viruses. These biosafety hurdles could be overcome by the use of recombinant single-cycle infectious arenaviruses. To explore this concept, we have developed a recombinant lymphocytic choriomeningitis virus (LCMV) (rLCMVΔGP/GFP) where we replaced the viral glycoprotein (GP) with the green fluorescent protein (GFP). We generated high titers of GP-pseudotyped rLCMVΔGP/GFP via genetic trans complementation using stable cell lines that constitutively express LCMV or LASV GPs. Replication of these GP-pseudotyped rLCMVΔGP/GFP viruses was restricted to GP-expressing cell lines. This system allowed us to rapidly and reliably characterize and quantify the neutralization activities of serum antibodies against LCMV and LASV within a BSL-2 facility. The sensitivity of the GFP-based microneutralization assay we developed was similar to that obtained with a conventionally used focus reduction neutralization (FRNT) assay. Using GP-pseudotyped rLCMVΔGP/GFP, we have also obtained evidence supporting the feasibility of this approach to identify and evaluate candidate antiviral drugs against HF arenaviruses without the need of BSL-4 laboratories.  相似文献   

13.
14.
The influenza A virus genome consists of eight RNA segments that associate with the viral polymerase proteins (PB1, PB2, and PA) and nucleoprotein (NP) to form ribonucleoprotein complexes (RNPs). The viral NS1 protein was previously shown to associate with these complexes, although it was not clear which RNP component mediated the interaction. Using individual TAP (tandem affinity purification)-tagged PB1, PB2, PA, and NP, we demonstrated that the NS1 protein interacts specifically with NP and not the polymerase subunits. The region of NS1 that binds NP was mapped to the RNA-binding domain.  相似文献   

15.
16.
The Old World arenavirus Lassa virus (LASV) is the causative agent of severe viral hemorrhagic fever (VHF) in humans and is the most prevalent human pathogen among arenaviruses. The present study investigated the largely unknown mechanisms of cell entry of LASV, a process know to be mediated solely by the virus envelope glycoprotein (GP). To circumvent biosafety restrictions associated with the use of live LASV, we used reverse genetics to generate a recombinant variant of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) expressing the LASV GP (rLCMV-LASVGP). The rescued rLCMV-LASVGP grew to titers comparable to that of LCMV and showed the receptor binding characteristics of LASV. We used rLCMV-LASVGP to characterize the cellular mechanisms of LASV entry in the context of a productive arenavirus infection. The kinetics of pH-dependent membrane fusion of rLCMV-LASVGP resembled those of the human-pathogenic New World arenavirus Junin virus (JUNV) and other enveloped viruses that use clathrin-mediated endocytosis for entry. However, rLCMV-LASVGP entered cells predominantly via a clathrin-, caveolin-, and dynamin-independent endocytotic pathway similar to the one recently described for LCMV. Productive infection of rLCMV-LASVGP was only mildly affected by a dominant negative mutant of Rab5 and was independent of Rab7, suggesting an unusual mechanism of delivery to endosomes. In addition, rLCMV-LASVGP infection was independent of actin but required intact microtubules. Our data indicate that LASV enters cells via a pathway distinct from the one used by human-pathogenic New World arenaviruses.  相似文献   

17.
汉坦病毒是引起肾综合征出血热(HFRS)和汉坦病毒型肺炎综合征(HPS)的主要病原体.其基因组由三节段的单股负链RNA组成,即S、M与L基因片段.汉坦病毒基因组的一个重要特点是每个基因片段的两个末端都有一段长18个核苷酸的高度保守的反向重复序列,互补可形成双链发夹结构,并且这一特点为不同型病毒所共有.为了研究该基因组末端保守的反向重复序列的功能,首先构建了汉坦病毒中国疫苗株Z10(汉滩型)及Z37(汉城型)的核蛋白原核表达载体,并在大肠杆菌中高效表达.经NI-NAT亲和柱和HiPrep16/10 DEAE离子交换柱液相色谱(FPLC)二步提纯,获得高纯度的重组核蛋白,并分别以胰蛋白酶消化后,用Western blotting进行区分和鉴定.以T4 DNA激酶同位素标记一对人工合成互补的18个核苷酸反向重复序列,制备双链探针.然后将该探针与纯化的Z10、Z37株的核蛋白NP进行非变性凝胶电泳迁移改变实验(EMSA)后发现,重组的Z10、Z37株的核蛋白NP,在体外均可特异地结合其基因组末端反向重复序列形成的双链探针.该结果表明,汉坦病毒基因组末端的反向重复序列是核蛋白重要结合位点,这对理解汉坦病毒核蛋白功能以及病毒复制过程中病毒粒子的包装机制有重要的意义.  相似文献   

18.
Several arenaviruses, chiefly Lassa virus (LASV), cause hemorrhagic fever (HF) disease in humans and pose a great public health concern in the regions in which they are endemic. Moreover, evidence indicates that the worldwide-distributed prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. The limited existing armamentarium to combat human-pathogenic arenaviruses underscores the importance of developing novel antiarenaviral drugs, a task that would be facilitated by the identification and characterization of virus-host cell factor interactions that contribute to the arenavirus life cycle. A genome-wide small interfering RNA (siRNA) screen identified sodium hydrogen exchanger 3 (NHE3) as required for efficient multiplication of LCMV in HeLa cells, but the mechanisms by which NHE activity contributed to the life cycle of LCMV remain unknown. Here we show that treatment with the NHE inhibitor 5-(N-ethyl-N-isopropyl) amiloride (EIPA) resulted in a robust inhibition of LCMV multiplication in both rodent (BHK-21) and human (A549) cells. EIPA-mediated inhibition was due not to interference with virus RNA replication, gene expression, or budding but rather to a blockade of virus cell entry. EIPA also inhibited cell entry mediated by the glycoproteins of the HF arenaviruses LASV and Junin virus (JUNV). Pharmacological and genetic studies revealed that cell entry of LCMV in A549 cells depended on actin remodeling and Pak1, suggesting a macropinocytosis-like cell entry pathway. Finally, zoniporide, an NHE inhibitor being explored as a therapeutic agent to treat myocardial infarction, inhibited LCMV propagation in culture cells. Our findings indicate that targeting NHEs could be a novel strategy to combat human-pathogenic arenaviruses.  相似文献   

19.
20.
The arenavirus small RING finger Z protein has been shown to be the main driving force of budding for several arenaviruses. This Z budding activity was found to be mediated by the late (L)-domain motifs P(T/S)AP and PPXY, located at the C terminus of Z. Here, we show that the Z protein of Tacaribe virus (TACV), a New World arenavirus, buds efficiently from cells despite lacking the canonical L-domain motifs P(T/S)AP and PPXY. Likewise, potential L-domain motifs ASAP and YLCL present in TACV Z did not exhibit any significant contribution to TACV Z budding activity. Budding of TACV Z was Tsg101 independent but required the activity of Vps4A/B. These results indicate that TACV Z utilizes a budding mechanism distinct from that reported for other arenaviruses.Arenaviruses are enveloped viruses with a bisegmented, negative-strand (NS) RNA genome and a life cycle restricted to the cell cytoplasm (1). Each RNA segment uses an ambisense coding strategy to direct the expression of two genes that are in opposite orientations and separated by a noncoding intergenic region. The large segment (7.2 kb) encodes the late (L) protein, an RNA-dependent RNA polymerase, and the small RING finger protein Z, which is the counterpart of the matrix (M) protein found in many enveloped NS RNA viruses. The small segment (3.5 kb) encodes the viral nucleoprotein (NP) and the glycoprotein precursor (GPC). The GPC is processed by the cellular protease S1P into GP1 and GP2 (13). Trimers of GP1/GP2 form the spikes that decorate the virus surface and mediate cell entry via receptor-mediated endocytosis (12).Arenaviruses merit significant interest both as tractable experimental model systems to study acute and persistent viral infections (18, 28) and as clinically important human pathogens. The Old World virus Lassa virus (LASV) and several New World (NW) arenaviruses cause hemorrhagic fever (HF) disease in humans, posing a serious public health problem (1). LASV is estimated to infect several hundred thousand individuals yearly in the regions of West Africa where it is endemic, resulting in a high number of Lassa fever (LF) cases associated with significant mortality and high morbidity. Notably, increased travel to and from regions of endemicity has led to the importation of LF into metropolitan areas, where the virus is not endemic, around the globe (11). Likewise, the NW arenavirus Junin virus causes Argentine HF, a severe illness with hemorrhagic and neurological manifestations and a fatality rate of 15 to 30% (7, 22, 27), while the NW Machupo and Guanarito arenaviruses have emerged as causative agents of HF in Bolivia and Venezuela, respectively (22). Public health concerns about HF arenavirus infections are exacerbated by the lack of licensed vaccines and by the fact that current antiarenavirus therapies are limited to the use of the nucleoside analogue ribavirin, which is only partially effective and is associated with significant side effects. Therefore, it is important to develop novel drugs to combat human pathogenic arenaviruses.Similarly to many other enveloped NS RNA viruses, arenavirus infectious particles bud from the plasma membranes of infected cells. Evidence indicates that the M proteins of many enveloped NS RNA viruses play critical roles in virus budding (3). Accordingly, many of these M proteins are, in the absence of any other virus polypeptide, competent in budding and can form virus-like particles (VLPs). Budding of M proteins is often directed by L-domain motifs, which most frequently correspond to one of the following sequences: P(T/S)AP, PPXY, YXXL, or FPIV (3). Efficient M-mediated budding requires interactions between viral L domains and host factors, many of them involved in the cellular multivesicular body (MVB) sorting pathway (3). MVB formation requires the activity of a network of cytoplasmic protein complexes known as endosomal sorting complexes required for transport (ESCRT). Tsg101 is a component of the ESCRT-1 complex and plays a key role in the biogenesis of MVB. An AAA ATPase, Vps4, which is present in humans as two isoforms, Vps4A and Vps4B, binds to components of ESCRT-3 and mediates dissociation of ESCRT-3 complex from the endosomal membrane. We (20, 26) and others (24) have documented that Z is the driving force of arenavirus budding. As with many other bona fide viral budding proteins, all known arenavirus Z proteins contain P(T/S)AP, PPXY, or both, L-domain motifs that play a critical role in Z budding (20, 24). Tacaribe virus (TACV) Z protein, however, constitutes a unique exception, as it lacks both P(T/S)AP and PPXY L-domain motifs (Fig. (Fig.1A1A).Open in a separate windowFIG. 1.Arenavirus Z protein and its budding efficiency. (A) Schematic representation of Z proteins from LCMV, LASV, and TACV. G corresponds to the strictly conserved glycine residue found at position 2 of all known arenavirus Z proteins. The locations of bona fide PTAP (LASV), PPPY (LASV and LCMV), and YXXL (LASV and TACV) L-domain motifs are indicated, as well as that of the L-like domain motifs STAP (LCMV) and ASAP (TACV). (B) Budding activity of TACV Z protein. 293T cells were transfected with 0.25 μg of either pC-TACV-Z-HA or pC-LASV-Z-HA. At 36 h posttransfection, tissue culture supernatants were collected, and VLPs and total cell lysates were prepared as described previously (2). Levels of Z proteins in VLPs and cell lysates were determined by WB using an antibody to HA (sc-7392; Santa Cruz). The budding efficiency of LASV Z was set at 1.0. The data are averages and standard deviations from three independent experiments.  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