首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到19条相似文献,搜索用时 93 毫秒
1.
禽流感(AvianInfluenza,AI)是由A型流感病毒所引起的各种家禽及野生禽类感染和/或疾病综合征[1]。根据其表面糖蛋白血凝素蛋白(Hemag glutinin,HA)和神经氨基酸酶(Neuraminidase,NA)的抗原关系不同,目前可分为16种HA亚型和9种NA亚型[2,3]。近几年来,南亚国家屡有禽流感病毒突破种间屏障作用,直接感染人类或其它哺乳动物,甚至致人死亡事件[4~6]的情况发生,因而赋予了禽流感全新的公共卫生学意义。因此,准确的了解和把握水禽,尤其是家养水禽的流感生态,对预防禽流感的发生具有非常重要的现实意义。为了防患于未然,近年来扬州大学农业…  相似文献   

2.
为了解H9N2亚型禽流感病毒(AIV)山东分离株的遗传变异情况,采用RT-PCR技术对16株从山东不同地区分离的H9N2亚型禽流感病毒的HA基因进行扩增、克隆和测序,并对所获得的HA全序列进行同源性和遗传进化分析。结果显示,16个分离株的裂解位点均为RSSR↓GLF,符合低致病性禽流感病毒的分子特征;有7~9个潜在糖基化位点;受体结合位点除198位有变异,其他位点均较保守;234位氨基酸均为L,具有与哺乳动物唾液酸α,2-6受体结合的特征;16个分离株HA基因核苷酸及氨基酸序列同源性分别为96.3%~99.9%和97.1%~99.6%;16个分离株同属于欧亚分支中的A/Duck/Hong Kong/Y280/97亚群。  相似文献   

3.
采用常规的血清学收验和特异性RT-PCR方法对华东地区家养水禽中流感病毒的带毒状况进行两年多的监测,分离鉴定出多株H6亚型禽流感病毒。对其中的一株A/Duck/Yangzhou/233/02(H6N2)(简称DkYZ23302)(H6N2)的表面膜蛋白基因进行了序列测定,并与GenBank中收录的其它序列进行了比较,遗传进化结果表明DkYZ23302的血凝素基因(HA)与近年香港分离的鸭源毒株DkHK346199(H6N1)、中国台湾鸡源毒株CkTaiwanna398的亲缘关系最近;而神经氨酸酶基因(NA)遗传进化分析结果表明DkYZ23302(H6N2)的NA基因起源于禽源H9N2亚型流感病毒,这可能是不同亚型禽流感病毒在水禽体内发生基因重配的结果。DkYZ23302(H6N2)的HA推导的氨基酸剪切位点序列为P-Q-I-E-T-R-D,为典型低致病性禽流感病毒的特征序列,与对SPF鸡的致病力试验相吻合。  相似文献   

4.
1998~2008年中国中部H9N2亚型AIV分离毒株HA基因的进化分析   总被引:1,自引:0,他引:1  
赵军  柴丽娜  王泽霖 《病毒学报》2011,27(2):122-128
从过去10年由中国中部分离的具有不同致病力的25株H9N2亚型禽流感选出6株(3#、12#、25#、14#、4#、22#)代表性毒株,利用RT-PCR扩增它们的HA基因,并比较分析该基因的序列,旨在探讨HA基因的变异对AIV毒力、抗原性变化的影响。结果表明:6株H9N2 AIV亚型分离株的HA基因在HA1和HA2的氨基酸裂解位点上没有出现高致病性禽流感病毒所特有的R-X-R/K-R模式,它们均为弱毒力毒株。HA上潜在糖基化位点除了3#和12#分离株多出一个之外,其余均为8个。3#和12#所表现出较强的致病性可能与其在HA的头部(HA1)的A抗原位点上多了一个糖基化位点(145~147aa),改变了HA基因空间构型有关,空间构型的改变导致抗HA抗体作用位点的变异或缺失并影响其较近的受体结合位点,从而改变该毒株的抗原性。研究结果提示需要持续跟踪H9N2 AIV在中国鸡群中的传播和进化,以便及时掌握疫情,有效防控禽流感。  相似文献   

5.
6.
从广东省疑似流感发病猪分离到1株H3N2亚型猪流感病毒(A/Swine/Guangdong/01/2005(H3N2)),对其各个基因进行克隆与测序,并与GenBank中收录的其它猪流感、禽流感和人流感的相关基因进行比较,结果表明,HA全基因与广东2003~2004年分离的H3N2猪流感毒株的核苷酸序列同源性在99%以上,与纽约90年代末分离的H3N2人流感毒株同源性在98.5%以上;NA基因与纽约1998~2000年分离的H3N2人流感毒株的核苷酸序列同源性在99%以上;NS基因、M基因的核苷酸序列与H1N1亚型猪流感毒株A/swine/HongKong/273/1994(H1N1)的核苷酸序列同源性较高,分别为97.9%、98.4%,与美洲A/swine/Iowa/17672/1988(H1N1)的核苷酸序列同源性分别为96.7%、97.1%;其他基因的核苷酸序列与H3N2人流感毒株具有很高的同源性。因此,推测其M和NS基因来源于H1N1亚型猪流感病毒,HA、NA及其他基因均来源于H3N2亚型人流感病毒。表明此H3N2亚型猪流感病毒为H3N2亚型人流感病毒和H1N1亚型猪流感病毒经基因重排而得到的重组病毒。  相似文献   

7.
禽流感病毒A/Chicken/Guangdong/SS/94(H9N2)HA基因的克隆及序列分析   总被引:10,自引:0,他引:10  
禽流感是由A型流感病毒引起的禽的一种疾病综合症.1878年首次在意大利爆发流行,当时称该病为"鸡瘟".之后,许多国家和地区都有该病的报道,包括美国、英国、澳大利亚、爱尔兰、比利时、荷兰、法国、俄罗斯、加拿大、以色列、匈牙利、日本、中国(包括香港)等[1-3].  相似文献   

8.
将LG1株H9N2亚型禽流感病毒在带有抗LG1株母源抗体的鸡胚中分4个独立系列连续传40代后,有3个系列从10~20代起在NA基因的#99位发生了可稳定遗传的碱基"G"到"A"的突变,并使氨基酸由蛋氨酸变为异亮氨酸;有2个系列从20~30代起在#473位发生了可稳定遗传的由"A"到"G"的碱基突变,导致相应的氨基酸由天冬酰胺变为丝氨酸,另一个传代系列在50代时也发生了同样的突变。在无抗体的鸡胚上的2个独立对照系列同样传了80代,在这2个位点没有发生突变,表明这2个突变与抗体的选择压相关。在抗LG1母源抗体阳性鸡胚的连续40代传代过程中,NA基因在有抗体组的四个传代系列碱基的非同义突变(NS)与同义突变(S)比为4.6(32/7),而在无抗体组NS/S比为2.0(16/8)。有抗体组NS/S值显著高于无抗体组,也显示出抗体的选择压作用。  相似文献   

9.
我国湖南地区因水禽家禽饲养较多且密集,是流感病毒高发省份。为了监测流感病毒的变异情况,采用RT-PCR技术扩增了10株H9N2亚型流感病毒的HA和NA基因,并进行了序列测定和分析。结果显示,10株分离株均属于欧亚分支中的Y280亚系;HA碱性裂解位点序列均为RSSR↓GLT,为低致病性流感病毒特征;HA受体结合位点234位均为L,具有与人唾液酸α-2,6受体结合的特性;NA蛋白均在颈部 (63–65位) 出现氨基酸缺失,这种缺失能增加流感病毒在哺乳动物中的复制能力。提示H9N2亚型毒株近年有毒力和致病力趋于转强的可能性,因此,要加强对H9N2亚型禽流感病毒的监测,密切关注它的重组趋势。  相似文献   

10.
两株H9N2亚型禽流行性病冒病毒HA基因序列分析   总被引:1,自引:0,他引:1  
程坚  刘红旗等 《病毒学报》2002,18(3):285-287
  相似文献   

11.
A purified antigen, HABA protein, has been derived from influenza virus concentrates by extraction with denaturing solvents. The protein lacks hemagglutinating activity but binds completely strain-specific, hemagglutination-inhibiting antibodies and induces neutralizing antibodies in experimental animals. Physicochemical characterization of HABA protein identifies it as a single homogeneous glycoprotein with a molecular weight of 78,000. On dissociation with guanidine or sodium dodecyl sulfate, in the presence of reducing agents, only one size of polypeptide with a molecular weight of the order of 40,000 is characteristic of the preparations. The data indicate that HABA protein is a dimer of HA(1) polypeptide of the influenza virus hemagglutinin substructure, and that only trace amounts of other polypeptides are present.  相似文献   

12.
The hemagglutinin, esterase, and fusion (HEF) glycoprotein of influenza C virus possesses receptor binding, receptor destroying, and membrane fusion activities. The HEF cDNAs from influenza C/Ann Arbor/1/50 (HEF-AA) and influenza C/Taylor/1223/47 (HEF-Tay) viruses were cloned and expressed, and transport of HEF to the cell surface was monitored by susceptibility to cleavage by exogenous trypsin, indirect immunofluorescence microscopy, and flow cytometry. Previously it has been found in studies with the C/Johannesburg/1/66 strain of influenza C virus (HEF-JHB) that transport of HEF to the cell surface is severely inhibited, and it is thought that the short cytoplasmic tail, Arg-Thr-Lys, is involved in blocking HEF cell surface expression (F. Oeffner, H.-D. Klenk, and G. Herrler, J. Gen. Virol. 80:363-369, 1999). As the cytoplasmic tail amino acid sequences of HEF-AA and HEF-Tay are identical to that of HEF-JHB, the data indicate that cell surface expression of HEF-AA and HEF-Tay is not inhibited by this amino acid sequence. Furthermore, the abundant cell surface transport of HEF-AA and HEF-Tay indicates that their cell surface expression does not require coexpression of another viral protein. The HEF-AA and HEF-Tay HEF glycoproteins bound human erythrocytes, promoted membrane fusion in a low-pH and trypsin-dependent manner, and displayed esterase activity, indicating that the HEF glycoprotein alone mediates all three known functions at the cell surface.  相似文献   

13.
Borna disease virus (BDV) is characterized by highly neurotropic infection. BDV enters its target cells using virus surface glycoprotein (G), but the cellular molecules mediating this process remain to be elucidated. We demonstrate here that the N-terminal product of G, GP1, interacts with the 78-kDa chaperone protein BiP. BiP was found at the surface of BDV-permissive cells, and anti-BiP antibody reduced BDV infection as well as GP1 binding to the cell surface. We also reveal that BiP localizes at the synapse of neurons. These results indicate that BiP may participate in the cell surface association of BDV.Borna disease virus (BDV) belongs to the Bornaviridae family of nonsegmented, negative-strand RNA viruses and is characterized by highly neurotropic and noncytopathic infection (18, 33). BDV infects a wide variety of host species and causes central nervous system (CNS) diseases in animals, which are frequently associated with behavioral disorders (14, 19, 29, 31). BDV cell entry is mediated by endocytosis, following the attachment of viral envelope glycoprotein (G) to the cellular receptor (2, 7, 8). BDV G is translated as a precursor protein, GP, which is posttranslationally cleaved by the cellular protease furin to generate two functional subunits of the N (GP1) and C (GP2) termini (28). Recent studies revealed that GP1 is involved in virus interaction with as-yet-unidentified cell surface receptor(s) and that GP2 mediates a pH-dependent fusion event between viral and cell membranes (2, 7, 27). In addition, a previous work using a hippocampal culture system suggested that BDV G is required for viral dissemination in neurons (2); however, cellular factors involved in BDV cell entry, especially cell surface association, remain to be elucidated.To extend our understanding of the role of BDV G in the interaction with the cell plasma membrane, we transfected GP1 fused with hemagglutinin-tobacco etch virus protease cleavage site-FLAG tags (GP1-TAP) into human oligodendroglioma OL cells. GP1-TAP was purified using anti-FLAG M2 affinity gel (Sigma). To verify that GP1-TAP binds to OL cells, the cells were incubated with 4 μg/ml GP1-TAP, and binding was detected by anti-FLAG M2 antibody (Sigma). A flow cytometric analysis indicated that GP1-TAP binds to OL cells (Fig. (Fig.1A).1A). To further validate the binding of GP1-TAP, we tested whether GP1-TAP inhibits BDV infection. OL cells were pretreated with 4 μg/ml GP1-TAP for 30 min. Proteins purified from mock-transfected cells using an anti-FLAG M2 affinity gel served as a control. The cells were then mixed with cell-free BDV. After 1 h of absorption, the supernatants were removed and fresh medium was added. At 3 days postinfection, the viral antigens were stained with anti-nucleoprotein (N) monoclonal and anti-matrix (M) polyclonal antibodies. As shown in Fig. Fig.1B,1B, GP1-TAP reduced BDV infection by 40% compared to levels for mock-treated cells. This result was consistent with earlier reports showing that recombinant GP1 protein binds to the cell surface and inhibits BDV infection (6, 20).Open in a separate windowFIG. 1.BDV GP1 binds to the cell surface. (A) Binding of BDV GP1 to OL cells. OL cells were incubated with GP1-TAP (solid line), and its binding was detected using anti-FLAG M2 antibody and flow cytometry. As a control, cells incubated with proteins purified from mock-transfected cells were detected by an anti-FLAG M2 antibody (dotted line). (B) Inhibition of BDV infection by GP1. OL cells pretreated with GP1-TAP were inoculated with the BDV huP2br strain. Values are the means + standard deviations (SD) from three independent experiments. **, P < 0.01.To investigate the host factor(s) that mediates the interaction of GP1 with the cell surface, a combination of tandem affinity purification (TAP) and liquid chromatography tandem mass spectrometry analyses was designed (13). We transfected GP1-TAP into OL cells and then purified GP1 from cell homogenates using a TAP strategy. We compared the purified proteins from the whole-cell and cytosol fractions (Fig. (Fig.2A),2A), and the bands detected only in the whole-cell fraction were determined as GP1-binding proteins in the membrane and/or nuclear fractions. In addition to GP1 protein (Fig. (Fig.2A,2A, arrow), we identified a specific band around 80 kDa in the whole-cell homogenate, but not in the cytosol fraction (Fig. (Fig.2A,2A, arrowhead), and determined that the band corresponded to the BiP (immunoglobulin heavy chain-binding protein) molecular chaperone, also called glucose-regulated protein 78 (GRP78), by mass spectrometry analysis. We confirmed the specific interaction between endogenous BiP and BDV G in infected cells by immunoprecipitation analysis (Fig. (Fig.2B).2B). To map the binding domain on BiP to GP1, we constructed a series of deletion mutants of the green fluorescent protein (GFP)-tagged BiP plasmid (Fig. (Fig.2C).2C). We transfected the mutant plasmids into BDV-infected OL cells and then performed an immunoprecipitation assay using anti-GFP antibody (Invitrogen). As shown in Fig. Fig.2D,2D, BDV G was coimmunoprecipitated with truncated BiP mutants, except for BiPΔN-GFP, which lacks the ATP-binding domain of BiP (lane 3), suggesting that BiP interacts with GP1 via its N-terminal region.Open in a separate windowFIG. 2.BDV GP1 interacts with BiP molecular chaperone. (A) TAP analysis of BDV GP1. Proteins coimmunoprecipitated with GP1-TAP in OL cells were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and visualized by silver staining. Cyt, cytosol fraction; Wc, whole-cell homogenate. Arrow, GP1-TAP; arrowhead, BiP. (B) Coimmunoprecipitation (IP) of BDV G and endogenous BiP. BDV G was immunoprecipitated from BDV-infected OL cells by anti-BDV G polyclonal antibody. Endogenous BiP was then detected by anti-BiP monoclonal antibody (Becton Dickinson). IgG, immunoglobulin G. (C) Schematic representation of deletion mutants of recombinant BiP-GFP. The known functional regions are indicated. (D) Immunoprecipitation analysis of BiP-GFP mutants in BDV-infected OL cells. The deletion plasmids were transfected and immunoprecipitated by anti-GFP antibody. Specific binding was detected using anti-BDV G antibody. Lane 1, GFP; lane 2, BiP-GFP; lane 3, BiPΔN-GFP; lane 4, BiPΔPB-GFP; lane 5, BiPΔC-GFP.BiP is known to be resident primarily in the endoplasmic reticulum and functions as a molecular chaperone involved in the folding process of nascent proteins, mostly through interaction with its peptide-binding domain (12, 17, 21). On the other hand, BiP has been reported to serve as a coreceptor of certain viruses at the plasma membrane (15, 34). Recent studies also revealed that cell surface BiP mediates the internalization of its ligands into cells (1, 10). We first investigated whether BiP is expressed on the cell surface of BDV-permissive OL and 293T cells using an anti-BiP polyclonal antibody (H-129; Santa Cruz Biotechnology, Inc.). As shown in Fig. Fig.3A,3A, BiP expression is detected on the surface of both cell lines. This result is in agreement with recent observations that BiP is expressed on the surface of various types of cells (9, 10, 15, 23, 24, 34). We also investigated whether BiP is expressed on the cell surface of BDV-nonpermissive cell lines, such as HeLa and CHO cells. As shown in Fig. Fig.3A,3A, we detected BiP expression on the surface of HeLa, but not CHO, cells. These observations were confirmed by immunofluorescence analysis (Fig. (Fig.3B).3B). Note that BiP is clearly detected at the endoplasmic reticulum in the permeabilized CHO cells by the antibody (see Fig. S1 in the supplemental material), suggesting that BiP is expressed at a very low level, if at all, on the surface of CHO cells. We next examined whether cell surface BiP serves as a binding molecule of BDV GP1. To test this, we performed an inhibition assay using an anti-BiP polyclonal antibody (N-20; Santa Cruz Biotechnology, Inc.) which recognizes the N terminus of BiP. As shown in Fig. Fig.3C,3C, the antibody inhibited GP1 binding to the cell surface by 40%. Furthermore, BDV infection was found to decrease by 70% when cells were treated with the antibody (Fig. (Fig.3D3D).Open in a separate windowFIG. 3.Cell surface BiP mediates cell association of BDV. (A) Flow cytometric analysis was performed with anti-BiP antibody (H-129) in BDV-permissive (OL and 293T) and -nonpermissive (HeLa and CHO) cells (solid lines). Cells stained with normal rabbit immunoglobulin G were used as a control (dotted lines). (B) Immunofluorescence analysis was performed by using anti-BiP antibody (H-129) with BDV-permissive and -nonpermissive cells. Arrows indicate BiP staining at the membrane. Scale bars, 10 μm. (C) Inhibition of GP1 binding by anti-BiP antibody (N-20). OL cells were pretreated with anti-BiP antibody, followed by labeling with GP1. GP1 binding on the cell surface was detected using flow cytometry. Values are the means + SD from three independent experiments. *, P < 0.05. (D) Inhibition of BDV infection by anti-BiP antibody. OL cells were incubated with 10 μg/ml anti-BiP antibody or normal goat immunoglobulin G and then the cells were mixed with cell-free BDV. After 1 h absorption, the supernatants were replaced with fresh medium. Virus infection was measured by immunofluorescence analysis using anti-N and -M antibodies at 3 days postinfection. Values are the means + SD from three independent experiments. *, P < 0.05. IgG, immunoglobulin G.To investigate the role of cell surface BiP in the infection of BDV, the BiP expression was inhibited by short interfering RNA (siRNA) in OL cells (see Fig. S2A in the supplemental material). We selected an siRNA (Hs_HSPA5_4; Qiagen, Inc.) which could partially downregulate the cell surface expression of BiP (see Fig. S2B in the supplemental material). However, siRNA treatment of BiP did not influence the infectivity of BDV in OL cells (see Fig. S2C in the supplemental material). This may be due to an incomplete reduction of BiP expression on the cell surface. Alternatively, while BiP mediates at least in part the cell surface association of BDV particles, this result may exhibit the presence of another, as-yet-unidentified BDV G-binding protein that is involved in the binding and subsequent cell entry of BDV.Previous studies demonstrated that BDV can be traced centripetally and transsynaptically after olfactory, ophthalmic, or intraperitoneal inoculation (3, 25). Migration of BDV to the CNS after footpad infection can be prevented by sciatic nerve transection (3). These observations suggest that BDV may disseminate primarily via neural networks. Recently, it has been demonstrated that BDV G was expressed at the termini of neurites or at contact sites of neurites (2), suggesting that local assembly of BDV may take place at the presynaptic terminals of synapses, similar to assembly of other neurotropic viruses (22, 26, 32). If BiP localizes at synapse sites, BiP may efficiently participate in the transmission of BDV particles at the synapses. To evaluate this hypothesis, we examined BiP localization in primary culture of mouse hippocampal neurons. After in vitro culture for 17 days, BiP localization was determined by an immunofluorescence assay without permeabilization. As shown in Fig. Fig.4A,4A, BiP signals were clearly detected at neurites, including the contact sites between dendrites and axons, as punctate staining (arrows), suggesting that BiP is expressed at the neuronal surface, most likely at the synapses. We next examined the localization of BiP with postsynaptic density 95 (PSD-95), a marker of postsynaptic density (5). Although BiP signals were detected mainly in the perinuclear area of the hippocampal neurons, punctate staining was also found at neurites colocalized with PSD-95 (Fig. (Fig.4B,4B, arrows). Taken together, these observations suggested that BiP is distributed at the synaptic surface, including the postsynaptic membrane, of neurons, a possible site for BDV budding and entry (2).Open in a separate windowFIG. 4.BiP localizes at the synaptic surface of hippocampus neurons. (A) Localization of BiP at synaptic surface. Hippocampal neurons were immunostained with anti-BiP antibody (N-20) without permeabilization. A differential interference contrast (DIC) image is shown. Dotted lines in the Merge panel indicate the dendrite outline. Arrows indicate BiP staining at the contact sites between axons and dendrites. (B) Colocalization between BiP and a postsynaptic protein. Hippocampal neurons were immunostained with anti-BiP (N-20) and anti-PSD-95 (Millipore) antibodies. Arrows indicate colocalized signals of BiP and PSD-95 at neurites. Scale bars, 10 μm.In summary, this study demonstrates that BiP is a GP1-binding protein at the synaptic surface. This is the first report showing the BDV G-binding factor on the cell surface. The first step of BDV entry might be mediated by the interaction of GP1 with as-yet-unidentified cell surface receptors, which may form a complex with other molecules, such as BiP. We showed that treatment with anti-BiP antibody affects BDV infection as well as GP1 binding to the cell surface (Fig. (Fig.3).3). Furthermore, synaptic distribution of BiP was found in hippocampal primary neurons (Fig. (Fig.4).4). These findings strongly suggest that BiP plays critical roles in BDV association with the neuronal surface via interaction with GP1. On the other hand, a BDV-nonpermissive cell line, HeLa, appeared to express BiP on the cell surface, suggesting that the cell surface BiP may not be necessarily involved in the infectivity of BDV. A recent study by Clemente et al. (6) revealed that following initial attachment to the cell surface, BDV is recruited to the plasma membrane lipid raft (LR) prior to internalization of the particles. The study suggested that BDV may use the LR as a platform to interact with additional host cell factor(s) required for efficient BDV internalization. Because BiP does not contain transmembrane regions, BiP needs another host protein(s) with transmembrane regions on the cell surface. It has been reported that cell surface BiP interacts with diverse proteins, such as major histocompatibility complex class I molecules (34), the voltage-dependent anion channel (9), and the DnaJ-like protein MTJ-1 (4), all of which associate with LR in the plasma membrane (16, 24, 35). Once BDV has attached to the cell surface, it might utilize such BiP-associated LR proteins for efficient cell surface attachment or internalization. Previously, it has been proposed that kainate 1 (KA-1) receptor might represent the BDV receptor within the CNS (11). Because some glutamate receptors are shown to bind to BiP (30), KA-1 receptors might interact with BiP and serve as a receptor complex for BDV. Further studies are required for a full understanding of the cell association processes, especially receptor binding, of BDV.   相似文献   

14.
2008年至2009年间,在湖南和湖北两省的活禽市场中分离到了14株H6亚型禽流感病毒,为了解这14株病毒之间的分子特征和差异,我们运用PCR和测序鉴定对这14株病毒的NA基因进行了分型,并对其表面基因HA和NA进行序列测定及序列分析.14株H6亚型病毒中,H6N2亚型12株,H6N6亚型2株.序列测定和进化分析结果显示:DK/HN/284的HA基因与其它13株的HA差异性较大,差异性达到19.4%~20.2%,其余13株毒同源性在94.2%~99.9%;N2亚型NA基因的同源性在91.1%~99.9%,差异性比较大;两株N6亚型NA基因同源性为89.5%,差异明显.这些数据表明:不同毒株呈现一定的地域性差异.与我国周边其它地区的H6亚型禽流感毒株序列进行比较发现,只有DK/HN/284的HA基因与香港早期的毒株可能有着共同的来源,其余都与香港和韩国等的毒株有着较大的差异性,并且各个毒株的HA基因上潜在的糖基化位点和受体结合位点也有所不同,这些数据表明,这些毒株表现出明显的异源性.  相似文献   

15.
16.
17.
New World arenaviruses, which cause severe hemorrhagic fever, rely upon their envelope glycoproteins for attachment and fusion into their host cell. Here we present the crystal structure of the Machupo virus GP1 attachment glycoprotein, which is responsible for high-affinity binding at the cell surface to the transferrin receptor. This first structure of an arenavirus glycoprotein shows that GP1 consists of a novel α/β fold. This provides a blueprint of the New World arenavirus attachment glycoproteins and reveals a new architecture of viral attachment, using a protein fold of unknown origins.Pathogenic human and animal viruses constitute a growing and persistent threat to global health (25). Machupo virus (MACV), responsible for Bolivian hemorrhagic fever (HF), is an apt example, being zoonotic and highly virulent. MACV was first isolated in 1963 and, along with Junín virus (JUNV), Guanarito virus (GTOV), Sabia virus (SABV), and Chapare virus (CAPV), comprises the HF viruses within clade B of the New World arenavirus family (13, 21). Clinical features of MACV infection during initial disease onset generally include fever, malaise, and headaches, developing over 7 to 10 days into severe HF (13). The high fatality rate (∼20%) and potential for global spread of this rodent-borne virus by deliberate dissemination have resulted in its classification by the National Institute for Allergy and Infectious Diseases as a high-priority category A biothreat agent (6).MACV is an ambisense RNA enveloped virus composed of a bisegmented genome. The L (large) segment encodes an RNA-dependent polymerase (L) and a zinc finger matrix protein (Z); the S (small) segment encodes the nucleoprotein (NP) and the viral glycoprotein precursor GPC (9). The L and NP proteins are coded in the conventional sense for a negative-sense RNA virus, while Z and GPC are transcribed in the opposite direction (Fig. (Fig.1).1). GPC is cleaved by the cellular proprotein convertase site 1 protease (39) to yield a stable complex composed of a 58-amino-acid signal peptide which is necessary for virus infectivity, a GP1 subunit which is involved in receptor attachment (199 amino acids), and a transmembrane-bound GP2 subunit (249 amino acids) which is putatively classified as a class I fusion protein (23, 38) (Fig. (Fig.11).Open in a separate windowFIG. 1.Schematic diagram of the ambisense, bisegmented arenavirus genome and details of the MACV GP1 sequence crystallized and ordered in the crystal structure. Both the L and S segments contain a central noncoding region (NCR). Arrows correspond to the coding directionality of the genes.MACV GP1 maintains low sequence identity with the GP1s of other New World HF arenaviruses (47, 27, 31, and 30% for JUNV, SABV, GTOV, and CAPV, respectively). Nevertheless, recent studies have shown that the transferrin receptor (TfR1) is a common cellular receptor for the GP1s of MACV, JUNV, GTOV, and SABV (24, 35, 36). These studies are an important step toward defining the viral tropism, and this interaction provides a target for the development of antivirals and prophylactic vaccines to prevent New World arenavirus infection. Knowledge of the molecular determinants of arenavirus attachment and fusion is a prerequisite for the rational development of immunotherapeutic and antiviral reagents (analogous to the development of neuraminidase inhibitors for the treatment of influenza [4]). To this end, we have solved the structure of the MACV GP1.The globular domain of MACV GP1 glycoprotein (MACV GP1) responsible for attachment to TfR1 (residues 87 to 257 from the complete mature GP1 which comprises residues 59 to 257; GenBank accession number AAS77647.1; cDNA synthesized by Codon Devices; Fig. Fig.1)1) was cloned into the pHLsec vector containing the chicken RPTPσ signal sequence (5). This region was selected based on the disorder predictions of RONN (44) and consideration of potential disulfide bond patterns. MACV GP1 was expressed in HEK 293T cells transfected with 2 mg DNA/liter of cell culture in the presence of 5 μM kifunensine, which prevents glycosylation processing, resulting in protein bearing oligomannose-type glycans (12). MACV GP1 protein was purified from the cell supernatant by using immobilized metal affinity followed by size-exclusion chromatography (SEC) using a Superdex 200 10/30 column (Amersham) equilibrated in 150 mM NaCl and 10 mM Tris, pH 8.0 (Fig. 2A and B). Protein yields were ∼2.0 mg MACV GP1/liter of cell culture. The binding activity of MACV GP1 for TfR1 (GenBank NC_BC001188, residues 122 to 760 cloned into the pHLsec vector [5]) was confirmed by coexpression and purification (as described above) of a MACV GP1-TfR1 complex from GlcNAc transferase I (GnTI)-deficient HEK 293S cells (37) (Fig. 2C and D).Open in a separate windowFIG. 2.Purification of MACV GP1 and MACV GP1-TfR1 complex. MACV GP1 and MACV GP1-TfR1 were expressed in HEK 293T (with 5 μM kifunensine) and GnTI-deficient HEK 293S cells, respectively. (A) SEC of MACV GP1 run on an S200 10/30 column. (B) A 4 to 12% gradient morpholineethanesulfonic acid-polyacrylamide gel electrophoresis assay of the resulting MACV GP1 from SEC run under reducing conditions (expected unglycosylated molecular mass, ∼22 kDa). The rightmost lane shows molecular mass markers. (C) SEC of MACV GP1-TfR1 complex run on an S200 10/30 column. Peak 1 corresponds to MACV GP1-TfR1 complex, and peak 2 corresponds to excess, unbound MACV GP1. (D) A 4 to 12% gradient morpholineethanesulfonic acid-polyacrylamide gel electrophoresis assay of the resulting protein from SEC run under reducing conditions. Lanes 1, 2, and 3 are consecutive fractions from peak 1, and lanes 4 and 5 are adjacent fractions from peak 2. The rightmost lane shows molecular mass markers. Note that peak 1 contains both GP1 (expected unglycosylated molecular mass, ∼22 kDa) and TfR1 (expected unglycosylated molecular mass, ∼71 kDa). Also, note that the apparent molecular mass difference observed between MACV GP1 in panel B and that in panel D is due to the different MACV GP1 glycoforms which result from expression in kifunensine-treated HEK 293T cells and GnTI-deficient HEK 293S cells.Purified MACV GP1 (concentrated to 12.5 mg/ml) crystallized from sitting drops of 100 nl plus 100 nl (25% [wt/vol] polyethylene glycol 3350, 0.2 M NaCl, and 0.1 M bis-Tris, pH 5.5) equilibrated against 95-μl reservoirs for 78 days at room temperature (42). Crystals were cryoprotected by immersion in reservoir solution plus 25% (vol/vol) glycerol and cryocooled in a 100 K gaseous nitrogen stream. X-ray diffraction data were recorded at the European Synchrotron Radiation Facility (ESRF), Grenoble, France. For phase determination, a crystal was soaked for 21 h with ∼10 mM potassium tetrachloroplatinate (II) and diffraction data were collected to a resolution of 3.4 Å on BM-14 at λ = 0.886 Å (the L1 edge for platinum), λ = 1.072 Å (inflection of the platinum L3 edge), and λ = 1.180 Å (low-energy remote) and on beamline ID14 EH1 at λ = 0.9334 Å (“high-energy remote” between platinum L1 and L2 edges). High-resolution (1.7-Å) data were recorded from a native crystal on ID14 EH1. Images were indexed, integrated, and scaled using HKL2000 (32). Data collection and crystallographic statistics are presented in Tables Tables11 and and22.

TABLE 1.

Data collection and phasing statistics for MACV GP1a,c
Data collectionNative GP1K2PtCl4
High-energy remotePeakInflectionLow-energy remote
BeamlineESRF ID14 EH1ESRF ID14 EH1ESRF BM14ESRF BM14ESRF BM14
Resolution (Å)50-1.7 (1.76-1.73)50-2.9 (2.99-2.90)50-3.4 (3.53-3.41)50-3.4 (3.55-3.43)50-3.4 (3.56-3.44)
Space groupP212121P212121P212121P212121P212121
Cell dimensions (Å)a = 51.5, b = 55.1, c = 61.3a = 52.0, b = 55.8, c = 60.9a = 52.0, b = 55.9, c = 61.0a = 52.0, b = 55.9, c = 61.2a = 52.0, b = 56.0, c = 61.2
Wavelength (Å)0.93340.93340.88561.0721.181
No. of unique reflections18,458 (843)4,315 (340)2,539 (248)2,539 (247)2,566 (246)
Completeness (%)98.2 (90.5)100.0 (100.0)96.8 (100.0)96.8 (100.0)97.6 (100.0)
Rmergeb0.11 (0.71)0.24 (0.72)0.17 (0.31)0.18 (0.44)0.23 (0.67)
II18.4 (3.5)12.0 (5.7)25.8 (12.5)15.3 (5.6)12.1 (3.6)
Avg redundancy14.1 (11.4)11.5 (12.0)13.4 (13.6)6.7 (6.8)6.7 (6.8)
Open in a separate windowaValues in parentheses refer to the relevant outer resolution shell.bRmerge = Σhkl Σi|I(hkl;i) − <I(hkl)>|/Σhkl ΣiI(hkl;i), where I(hkl;i) is the intensity of an individual measurement and <I(hkl)> is the average intensity from multiple observations.cMultiple-wavelength anomalous dispersion phasing statistics were as follows. SHARP figures of merit for centrics were 0.735 (41 to 10 Å), 0.241 (41 to 3 Å), and 0.072 (3.1 to 3.0 Å). SHARP figures of merit for acentrics were 0.913 (41 to 10 Å), 0.347 (41 to 3 Å), and 0.143 (3.1 to 3.0 Å). SHARP phasing power (isomorphous/anomalous) was as follows: high remote, −/0.73; peak, 0.15/1.07; inflection, 0.44/0.57; low remote, 0.29/0.21. Solvent-flattened figures of merit were 0.818 (41 to 11 Å), 0.789 (41 to 4.0 Å), and 0.767 (4.1 to 4.0 Å).

TABLE 2.

Refinement statistics for MACV GP1a
RefinementNative GP1
Resolution range (Å)32-1.7 (1.78-1.73)
No. of reflections17,465 (1,163)
Rfactor (%)b17.2 (23.0)
Rfree (%)c21.3 (30.5)
RMSDd bonds (Å)0.012
RMSD angles (°)1.4
Atoms per asymmetric unit (protein/water/sugar)1,239/198/70
Avg B-factors (protein/water/sugar) (Å2)16.0, 23.1, 28.5
Open in a separate windowaValues in parentheses refer to the relevant outer resolution shell.bRfactor = ΣhklFobs| − k|Fcalc‖/Σhkl|Fobs|.cRfree equals the Rfactor as calculated above against 5% of the data not used during refinement.dRMSD, root mean square deviation from ideal geometry.Phase determination used the multiple-wavelength anomalous dispersion method. Heavy atom positions were identified using SHELXD (40) and refined using SHARP (20), solvent flattening was performed using SOLOMON (2) and DM (17), and the resulting low-resolution (4-Å) electron density map was used for initial model building of the single molecule in the asymmetric unit (solvent content, 50%). This initial model was placed into the high-resolution data using PHASER (30), and model building was completed automatically using ARP-wARP (33). Structure refinement iterated cycles of restrained refinement with TLS using REFMAC5 (31) and manual rebuilding using COOT (22). The final model was validated using MolProbity (19). Of the residues, 96.7% lie within favored regions of the Ramachandran plot and the remainder lie in additionally allowed regions (19).The 1.7-Å resolution structure of MACV GP1 is shown in Fig. Fig.3.3. Although the MACV GP1 used for crystallization included residues 87 to 257 (Fig. (Fig.1),1), we do not see electron density for the last 20 C-terminal residues (the residues preceding the point of GP1/GP2 cleavage). MACV GP1 appears to be monomeric in the crystal. Structural database comparison (27, 45) suggests that MACV GP1 is composed of a fold that has not been previously observed. The N and C termini extend in the same direction, and the secondary structure consists of seven antiparallel β-strands forming a left-handed sheet (designated 1 to 7); three α-helices, one of which is preceded by a 310 helix; and two additional 310 helices (Fig. 3A and B; see also Fig. S1 in the supplemental material). When looking onto the edge of the β-sheet, the overall fold appears to be similar to the shape of a positive meniscus lens where the three large helices protect the convex side of the β-sheet whereas the concave side of the β-sheet is largely uncovered (Fig. 3B and D). The secondary structure is stabilized by four disulfide bonds which are present around the plane of the lens (Fig. (Fig.3C).3C). Two of these disulfide bonds appear to be conserved across the New World HF arenaviruses, a third is also found in the GP1 of JUNV, while the fourth stabilizes a MACV-specific insertion (Fig. 3C to E). The presence of an additional conserved disulfide bond between MACV GP1 and JUNV GP1 reflects the close relationship between these two viruses with respect to other New World arenaviruses (8, 11, 14); thus, for the portion of the GP1 that we have analyzed, the sequence identity is 40% (Fig. (Fig.3E),3E), and we would therefore expect the two structures to agree with approximately 1.2-Å root mean square deviation over matching Cα atoms (16).Open in a separate windowFIG. 3.Structure of MACV GP1. (A) Cartoon diagram of MACV GP1 colored as a rainbow with the N terminus shown in blue and the C terminus in red. The N and C termini are marked with blue and red spheres, respectively. (B) View of MACV GP1 rotated by 90° from that in panel A. (C) Cα trace of MACV GP1 colored gray with carbohydrate moieties (GlcNAc) observed at N-linked glycosylation sites (nitrogen atoms colored blue, oxygen atoms colored red, and carbon atoms colored yellow) and disulfide bonds (carbon atoms colored gray and sulfur atoms colored gold) shown as sticks. Disulfide bond pairs are numbered in magenta according to panel E. (D) View of MACV GP1 rotated by 90° from that in panel C. (E) Sequence alignment of residues observed in the MACV GP1 crystal structure with the GP1s of JUNV, GTOV, SABV, and CAPV (determined by ClustalW [15], plotted by ESPRIPT [26], and adjusted by hand). Secondary structure elements are shown with an arrow (β-strand, β1 to -7), and helices are shown as spirals, with α-helices shown in bold (α-helix, α1 to -3; 310 helix, η1 to -3). Residues which are highlighted red are fully conserved, residues which are colored red are partially conserved, and residues which are black are not conserved. Residues which are solvent accessible (as determined by ESPRIPT [26]) are highlighted by bars below the sequence and are colored light blue (partially accessible) or dark blue (fully accessible). Conserved amino acids which are solvent exposed are annotated below the alignment with yellow spheres. Amino acids which correspond to predicted N-linked glycosylation sites are marked with blue boxes and annotated beneath the alignment as yellow (conserved across all sequences) and green (not conserved) spheres. The bottom row of the alignment marks the disulfide bond pairs as marked in panel C.Four N-linked glycosylation sites lie on the perimeter of the plane of the β-sheet (Fig. 3C and D). Electron density was observed for at least one N-acetylglucosamine (GlcNAc) residue at each of these sites (Fig. (Fig.4).4). B-factors for glycan atoms (Table (Table2)2) are not significantly greater than those of other surface atoms. Deglycosylation of MACV GP1 with endo F1 resulted in precipitation (data not shown), suggesting that, as reported for some other systems (34), glycans solubilize the protein. Protein-carbohydrate interactions were observed for several glycans. For example, GlcNAc at Asn95 forms a classic stacking interaction between the side chain of Phe98 and the pyranose ring (Fig. (Fig.4A)4A) (7, 34, 43) (the rest of the glycan projects across a protein surface which bears further solvent-exposed aromatic residues). The glycan at Asn178 lies at the center of a cavity on the protein surface, braced by a network of hydrogen bonds (Fig. (Fig.4C),4C), including one to Glu184 which may stabilize the loop following the β6 strand. Oligomannose structures are often observed on glycoproteins from enveloped viruses, for example, human immunodeficiency virus type 1, dengue virus, and Ebola virus (3, 18, 29, 41), where they can influence viral tropism and stimulate the host immune response (28). While glycosylation processing of complex-type structures is influenced by tissue-specific processing, oligomannose-type glycans are frequently highly conserved between recombinant material and infectious virions. For example, the oligomannose-type glycans of human immunodeficiency virus type 1 gp120 are also present in recombinant gp120 monomers expressed in CHO cells (46). However, analysis of N-linked glycosylation by matrix-assisted laser desorption ionization-time of flight mass spectrometry (Fig. (Fig.4D;4D; see also Table S1 in the supplemental material) demonstrated that recombinant MACV GP1 contains instead highly branched, extensively heterogeneous, complex-type glycans when expressed in the absence of any glycosidase inhibitors.Open in a separate windowFIG. 4.Analysis of N-linked glycans on MACV GP1. Enlarged view of the GlcNAc residues at Asn95 (A), Asn137 and Asn166 (B), and Asn178 (C). MACV GP1 is shown as a cartoon representation with asparagines and GlcNAc structures shown as sticks. Carbon atoms of the GlcNAc moieties are shown in yellow, and those of asparagine side chains are gray. Dashed lines correspond to hydrogen bonds between GlcNAc moieties and amino acids. A maximum likelihood weighted 2Fo-Fc electron density map was calculated using the final refined model and is displayed around the GlcNAc residues contoured at 1σ. (D) Mass spectrometric analyses of MACV GP1. Matrix-assisted laser desorption ionization-time of flight mass spectrometry of desialylated N-linked glycans ([M + Na]+ ions) released from MACV GP1 expressed in HEK 293T cells. Symbols used for the structural formulae are as follows: ⋄, Gal; ⧫, GalNAc; ▪, GlcNAc; ○, Man; ·⃟, Fuc. The linkage position is shown by the angle of the lines linking the sugar residues (vertical line, 2-link; forward slash, 3-link; horizontal line, 4-link; backward slash, 6-link). Full annotation of the spectra, together with the anomericity indicated by full lines for β bonds and broken lines for α bonds, is provided in Table S1 in the supplemental material.Analyses of N-linked glycosylation sites across our GP1 segment from all New World HF arenaviruses (MACV, JUNV, GTOV, SABV, and CAPV) reveal a total of 10 potential sites which, when mapped onto the structure of MACV GP1, decorate solvent-accessible loops on the perimeter of the β-sheet (Fig. (Fig.5A).5A). Two sites are completely conserved, an additional site is conserved between MACV and JUNV, and the majority of glycans cluster at a specific side of the sheet (Fig. (Fig.5A).5A). In contrast, solvent-accessible residues completely conserved across the New World HF arenaviruses are distributed in an almost complementary pattern across the surface (Fig. (Fig.5B).5B). We suggest that the area containing the carbohydrate cluster is likely to be structurally variable among New World HF arenaviruses and that surfaces involved in receptor binding and interaction with GP2 are likely to lie outside this region, perhaps focused on the regions of the surface containing the conserved residues (Fig. (Fig.5B5B).Open in a separate windowFIG. 5.Conservation of amino acid sequence and N-linked glycosylation sites across New World HF arenaviruses mapped onto the structure of MACV GP1 (gray cartoon). (A) N-linked glycosylation sites from MACV GP1, JUNV GP1, GTOV GP1, SABV GP1, and CAPV GP1 mapped as spheres on the structure of MACV GP1. Spheres colored yellow are conserved across all viruses, and spheres colored green are not conserved. Residues that are clearly visible are labeled according to residue number. (B) Conserved amino acids which are solvent exposed (calculated by ESPRIPT [26]) (Fig. (Fig.3E)3E) from MACV GP1, JUNV GP1, GTOV GP1, SABV GP1, and CAPV GP1 mapped as spheres on the structure of MACV GP1. Spheres colored orange correspond to residues conserved across all New World HF arenaviruses.The crystal structure of the MACV GP1 reported herein represents the first structure of a New World arenavirus GP1. In agreement with previous phylogenetic studies (8, 11, 14), our structure-based analysis of conserved disulfide bonds and predicted N-linked glycosylation suggests that MACV GP1 is likely to have the greatest structural similarity to the JUNV GP1. More generally, given the conserved tropism of both pathogenic and nonpathogenic New World clade B arenaviruses for TfR1-expressing cells (1), we propose that this new protein fold defines the architecture of New World arenavirus attachment glycoproteins. The origins of this protein fold are unknown; it bears no relation to the host ligand of TfR1, transferrin. However, we note that the GPC gene is antisense, if we consider the Arenaviridae to be aberrant negative-sense RNA viruses. We suggest that either the antisense protein GP1 may have been recruited from the host (although the structure of GP1 does not resemble any seen so far in a host protein) or it may have originated de novo relatively recently, which might explain the large divergence in sequence between GP1s from different viral isolates (10).  相似文献   

18.
Human influenza is a seasonal disease associated with significant morbidity and mortality. The most effective means for controlling infection and thereby reducing morbidity and mortality is vaccination with a three inactivated influenza virus strains mixture, or by intranasal administration of a group of three different live attenuated influenza vaccine strains. Comparing to the inactivated vaccine, the attenuated live viruses allow better elicitation of a long-lasting and broader immune (humoral and cellular) response that represents a naturally occurring transient infection. The cold-adapted (ca) influenza A/AA/6/60 (H2N2) (AA ca) virus is the backbone for the live attenuated trivalent seasonal influenza vaccine licensed in the United States. Similarly, the influenza A components of live-attenuated vaccines used in Russia have been prepared as reassortants of the cold-adapted (ca) H2N2 viruses, A/Leningrad/134/17/57-ca (Len/17) and A/Leningrad/134/47/57-ca (Len/47) along with virulent epidemic strains. However, the mechanism of temperature-sensitive attenuation is largely elusive. To understand how modification at genetic level of influenza virus would result in attenuation of human influenza virus A/PR/8/34 (H1N1,A/PR8), we investigated the involvement of key mutations in the PB1 and/or PB2 genes in attenuation of influenza virus in vitro and in vivo. We have demonstrated that a few of residues in PB1 and PB2 are critical for the phenotypes of live attenuated, temperature sensitive influenza viruses by minigenome assay and real-time PCR. The information of these mutation loci could be used for elucidation of mechanism of temperature-sensitive attenuation and as a new strategy for influenza vaccine development.  相似文献   

19.
为揭示广东地区2007~2010年甲型H3N2毒株血凝素(HA)基因特征和变异,采用时空抽样方法抽样,检测广东2007~2010年甲型H3N2毒株HA基因核苷酸序列,同时检索全球HA基因序列作为对照,采用Lasergene 7.1和Mega 5.05软件对HA基因核苷酸序列进行比对和分析;并结合流行病学资料,对变异毒株进行进化速度分析;同时进行抗原分析。结果发现,广东2007~2010年H3N2毒株HA基因同义进化(Ks)和错义进化(Ka)速度分别为2.06×1E-3~2.23×1E-3核苷酸/年和1.05×1E-3~1.21×1E-3核苷酸/年,HA1较HA2的错义突变速率要高3.13倍。与疫苗株A/Perth/16/2009的HA基因比较,2009年广东毒株同源性达到98.8%~99.7%、2010年同源性达到98.0%~98.4%。在广东2007~2010年毒株中,HA1五个抗原表位均有氨基酸位点变异,尤其是2010年毒株B区(N160K)和D区(K174R/N)的变异;此外,广东2010年毒株受体结合部位(RBS)还发生K189E/N/Q和T228A置换变异;两个糖基化位点变异影响到抗原性;目前使用的H3N2疫苗株与目前流行毒株的抗原性有差异。广东地区2007~2010年的毒株中,血凝抑制抗体的抗原分析结果有差异。结果提示,目前广东乃至全球甲型H3N2毒株HA1B区和D区均有氨基酸位点变异,RBS的两个位点发生置换,糖基化位点变异影响到表位A区和B区抗原性;与WHO推荐2011年流感H3N2毒株疫苗株比较,目前流行毒株HA基因有抗原位点变异。  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