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 62 毫秒
1.
The human parainfluenza virus type 3 (HPIV3) fusion (F) and hemagglutinin-neuraminidase (HN) glycoproteins are the principal components involved in virion receptor binding, membrane penetration, and ultimately, syncytium formation. While the requirement for both F and HN in this process has been determined from recombinant expression studies, stable physical association of these proteins in coimmunoprecipitation studies has not been observed. In addition, coexpression of other heterologous paramyxovirus F or HN glycoproteins with either HPIV3 F or HN does not result in the formation of syncytia, suggesting serotype-specific protein differences. In this study, we report that simian virus 5 and Sendai virus heterologous HN proteins and measles virus hemagglutinin (H) were found to be down-regulated when coexpressed with HPIV3 F. As an alternative to detecting physical associations of these proteins by coimmunoprecipitation, further studies were performed with a mutant HPIV3 F protein (F-KDEL) lacking a transmembrane anchor and cytoplasmic tail and containing a carboxyl-terminal retention signal for the endoplasmic reticulum (ER). F-KDEL was defective for transport to the cell surface and could down-regulate surface expression of HPIV3 HN and heterologous HN/H proteins from simian virus 5, Sendai virus, and measles virus in coexpression experiments. HN/H down-regulation appeared to result, in part, from an early block to HPIV3 HN synthesis, as well as an instability of the heterologous HN/H proteins within the ER. In contrast, coexpression of F-KDEL with HPIV3 wild-type F or the heterologous receptor-binding proteins, respiratory syncytial virus glycoprotein (G) and vesicular stomatitis virus glycoprotein (G), were not affected in transport to the cell surface. Together, these results support the notion that the reported serotype-specific restriction of syncytium formation may involve, in part, down-regulation of heterologous HN expression.  相似文献   

2.
cDNAs encoding the G glycoprotein of respiratory syncytial virus and the hemagglutinin-neuraminidase (HN) glycoprotein of parainfluenza virus type 3 were modified by site-specific mutagenesis and restriction fragment replacement to encode chimeric proteins consisting of the cytoplasmic and transmembrane domains of one protein fused to the ectodomain of the other. In the case of the HN ectodomain attached to the G transmembrane and cytoplasmic domains, cell surface expression of the chimera was reduced. Otherwise, the presence of the heterologous transmembrane and cytoplasmic domains had little effect on the processing of the HN or G ectodomain, as assayed by the acquisition of N-linked and O-linked carbohydrates, transport to the cell surface and, in the case of HN, folding, oligomerization, and hemadsorption activity. These results showed that the synthesis and processing of each ectodomain did not require the homologous transmembrane and cytoplasmic domains. In particular, O glycosylation of the G protein was specified fully by its ectodomain, even though this domain is highly divergent among the respiratory syncytial virus antigenic subgroups. In addition, whereas the cytoplasmic and transmembrane domains of the G protein were relatively highly conserved, they were nonetheless fully replaceable without significantly affecting processing.  相似文献   

3.
In the assembly of paramyxoviruses, interactions between viral proteins are presumed to be specific. The focus of this study is to elucidate the protein-protein interactions during the final stage of viral assembly that result in the incorporation of the viral envelope proteins into virions. To this end, we examined the specificity of HN incorporation into progeny virions by transiently transfecting HN cDNA genes into Sendai virus (SV)-infected cells. SV HN expressed from cDNA was efficiently incorporated into progeny Sendai virions, whereas Newcastle disease virus (NDV) HN was not. This observation supports the theory of a selective mechanism for HN incorporation. To identify the region on HN responsible for the selective incorporation, we constructed chimeric SV and NDV HN cDNAs and evaluated the incorporation of expressed proteins into progeny virions. Chimera HN that contained the SV cytoplasmic domain fused to the transmembrane and external domains of the NDV HN was incorporated to SV particles, indicating that amino acids in the cytoplasmic domain are responsible for the observed specificity. Additional experiments using the chimeric HNs showed that 14 N-terminal amino acids are sufficient for the specificity. Further analysis identified five consecutive amino acids (residues 10 to 14) that were required for the specific incorporation of HN into SV. These residues are conserved among all strains of SV as well as those of its counterpart, human parainfluenza virus type 1. These results suggest that this region near the N terminus of HN interacts with another viral protein(s) to lead to the specific incorporation of HN into progeny virions.  相似文献   

4.
Previously, we showed that Sendai virus fusion protein (F) acts as an inhibitor of neuraminidase activity of hemagglutinin-neuraminidase (HN) protein. Here we report that synthetic peptides derived from the heptad repeat region proximal to the transmembrane domain (HR2) of Sendai virus F inhibit fusion and enhance the enzymatic activity of the HN. This occurs on the virus-bound HN and on its soluble globular head. The enhancing effect on virus-bound HN is reversible and depends on the presence of F. The data indicate that, by binding to the HN ectodomain, the HR2 peptides abolish the F inhibition of HN and disrupt the communication between the F and HN essential to promote virus-cell fusion.  相似文献   

5.
The role of specific sequences in the transmembrane (TM) domain of Newcastle disease virus (NDV) fusion (F) protein in the structure and function of this protein was assessed by replacing this domain with the F protein TM domains from two other paramyxoviruses, Sendai virus (SV) and measles virus (MV), or the TM domain of the unrelated glycoprotein (G) of vesicular stomatitis virus (VSV). Mutant proteins with the SV or MV F protein TM domains were expressed, transported to cell surfaces, and proteolytically cleaved at levels comparable to that of the wild-type protein, while mutant proteins with the VSV G protein TM domain were less efficiently expressed on cell surfaces and proteolytically cleaved. All mutant proteins were defective in all steps of membrane fusion, including hemifusion. In contrast to the wild-type protein, the mutant proteins did not form detectable complexes with the NDV hemagglutinin-neuraminidase (HN) protein. As determined by binding of conformation-sensitive antibodies, the conformations of the ectodomains of the mutant proteins were altered. These results show that the specific sequence of the TM domain of the NDV F protein is important for the conformation of the preactivation form of the ectodomain, the interactions of the protein with HN protein, and fusion activity.  相似文献   

6.
Q Yao  R W Compans 《Journal of virology》1995,69(11):7045-7053
We have investigated the roles of the cytoplasmic domains of the human parainfluenza virus type 2 (PI2) and type 3 (PI3) fusion (F) proteins in protein transport and cell fusion activity. By using the vaccinia virus-T7 transient expression system, a series of F protein cytoplasmic tail truncation mutants was studied with respect to intracellular and surface expression and the ability to induce cell fusion when coexpressed with the corresponding hemagglutinin-neuraminidase (HN) proteins. All of the cytoplasmic tail truncation mutants of PI2F were expressed at high levels intracellularly or on cell surfaces as measured by immunoprecipitation and cell surface biotinylation assays. In addition, when coexpressed with PI2HN, these truncation mutants of PI2F were all found to be essentially unimpaired in the ability to induce cell fusion as measured by a quantitative cell fusion assay. In contrast, surface expression and cell fusion activity were found to be eliminated by a mutant of PI3F in which the entire cytoplasmic tail was deleted, and the mutant protein appeared to be unable to assemble into a high-molecular-weight oligomeric structure. To further investigate whether there is a specific sequence requirement in the cytoplasmic tail of PI3F, a chimeric protein consisting of the PI3F extracellular and transmembrane domains and the PI2F cytoplasmic tail was constructed. This chimeric protein was detected on the surface, and it was capable of inducing cell fusion when expressed together with PI3HN, although the fusogenic activity was reduced compared with that of wild-type PI3F. These results demonstrate that although PI2 and PI3 viruses belong to the same parainfluenza virus genus, these viruses show marked differences with respect to functional requirements for the cytoplasmic tail of the F glycoprotein.  相似文献   

7.
We describe the development of novel lentivirus vectors based on simian immunodeficiency virus from African green monkey (SIVagm) pseudotyped with Sendai virus (SeV) envelope glycoproteins. SeV fusion (F) and hemagglutinin-neuraminidase (HN) proteins were successfully incorporated into the SIVagm-based vector by truncation of the cytoplasmic tail of the F protein and by addition of the cytoplasmic tail of SIVagm transmembrane envelope protein to the N terminus of the HN protein. As with the vesicular stomatitis virus G glycoprotein-pseudotyped vector, the mutant SeV F- and HN-pseudotyped SIVagm vector was able to transduce various types of animal and human cell lines. Furthermore, the vector was able to transduce an enhanced green fluorescent protein reporter gene into polarized epithelial cells of rat trachea from the apical and basolateral sides. Therefore, SeV F- and HN-pseudotyped SIVagm vectors have considerable potential for effective use in gene therapy for various therapies, including respiratory diseases.  相似文献   

8.
Enveloped viruses contain glycoproteins protruding from the viral membrane. These proteins play a crucial role in the extra-cellular steps of the virus life cycle, namely attachment to and entry into cells. Their role during the intracellular late phase of virus multiplication has been less appreciated, overlooked by the documented central organizer role of the matrix M protein. Sendai virus, a member of the Paramyxoviridae family, expresses two trans-membrane proteins on its surface, HN and F. In previous work, we have shown that suppression of F in the context of an infection, results in about 70% reduction of virus particle production, a reduction similar to that observed upon suppression of the matrix M protein. Moreover, a TYTLE motif present in F cytoplasmic tail has been proposed essential for virus particle production. In the present work, using original alternate conditional siRNA suppression systems, we generated a double F gene recombinant Sendai virus expressing wt-F and a nonviable mutated TYTLE/5A F protein (F5A). Suppression of the wild type F gene expression in cells infected with this virus allowed the analysis of F5A properties in the context of the infection. Coupling confocal imaging analysis to biochemical characterization, we found that F5A i) was not expressed at the cell surface but restricted to the endoplasmic reticulum, ii) was still capable of interaction with M and iii) had profound effect on M and HN cellular distribution. On the basis of these data, we propose a model for SeV particle formation based on an M/F complex that would serve as nucleation site for virus particle assembly at the cell surface.  相似文献   

9.
Paramyxoviruses cause a wide variety of human and animal diseases. They infect host cells using the coordinated action of two surface glycoproteins, the receptor binding protein (HN, H, or G) and the fusion protein (F). HN binds sialic acid on host cells (hemagglutinin activity) and hydrolyzes these receptors during viral egress (neuraminidase activity, NA). Additionally, receptor binding is thought to induce a conformational change in HN that subsequently triggers major refolding in homotypic F, resulting in fusion of virus and target cell membranes. HN is an oligomeric type II transmembrane protein with a short cytoplasmic domain and a large ectodomain comprising a long helical stalk and large globular head domain containing the enzymatic functions (NA domain). Extensive biochemical characterization has revealed that HN-stalk residues determine F specificity and activation. However, the F/HN interaction and the mechanisms whereby receptor binding regulates F activation are poorly defined. Recently, a structure of Newcastle disease virus (NDV) HN ectodomain revealed the heads (NA domains) in a “4-heads-down” conformation whereby two of the heads form a symmetrical interaction with two sides of the stalk. The interface includes stalk residues implicated in triggering F, and the heads sterically shield these residues from interaction with F (at least on two sides). Here we report the x-ray crystal structure of parainfluenza virus 5 (PIV5) HN ectodomain in a “2-heads-up/2-heads-down” conformation where two heads (covalent dimers) are in the “down position,” forming a similar interface as observed in the NDV HN ectodomain structure, and two heads are in an “up position.” The structure supports a model in which the heads of HN transition from down to up upon receptor binding thereby releasing steric constraints and facilitating the interaction between critical HN-stalk residues and F.  相似文献   

10.
Interactions between viral glycoproteins, matrix protein and nucleocapsid sustain assembly of parainfluenza viruses at the plasma membrane. Although the protein interactions required for virion formation are considered to be highly specific, virions lacking envelope glycoprotein(s) can be produced, thus the molecular interactions driving viral assembly and production are still unclear. Sendai virus (SeV) and human parainfluenza virus type 1 (hPIV1) are highly similar in structure, however, the cytoplasmic tail sequences of the envelope glycoproteins (HN and F) are relatively less conserved. To unveil the specific role of the envelope glycoproteins in viral assembly, we created chimeric SeVs whose HN (rSeVhHN) or HN and F (rSeVh(HN+F)) were replaced with those of hPIV1. rSeVhHN grew as efficiently as wt SeV or hPIV1, suggesting that the sequence difference in HN does not have a significant impact on SeV replication and virion production. In sharp contrast, the growth of rSeVh(HN+F) was significantly impaired compared to rSeVhHN. rSeVh(HN+Fstail) which expresses a chimeric hPIV1 F with the SeV cytoplasmic tail sequence grew similar to wt SeV or rSeVhHN. Further analysis indicated that the F cytoplasmic tail plays a critical role in cell surface expression/accumulation of HN and F, as well as NP and M association at the plasma membrane. Trafficking of nucelocapsids in infected cells was not significantly affected by the origin of F, suggesting that F cytoplasmic tail is not involved in intracellular movement. These results demonstrate the role of the F cytoplasmic tail in accumulation of structural components at the plasma membrane assembly sites.  相似文献   

11.
The cell fusion activity of most paramyxoviruses requires coexpression of a fusion protein (F) and a hemagglutinin-neuraminidase protein (HN) which are derived from the same virus type. To define the domain of the HN protein which interacts with the F protein in a type-specific manner a series of chimeric HN proteins between two different paramyxoviruses, Sendai virus (SN) and human parainfluenza virus type 3 (PI3), was constructed and coexpressed with the SN-F protein by using the vaccinia virus T7 RNA polymerase transient-expression system. Quantitative assays were used to evaluate cell surface expression as well as fusion-promoting activities of the chimeric HN molecules. A chimeric HN protein [SN(140)] containing 140 N-terminal amino acids derived from SN-HN and the remainder (432 amino acids) derived from PI3-HN was found to promote cell fusion with the SN-F protein. In contrast, a second chimeric HN with 137 amino acids from SN-HN at the N terminus could not promote fusion with SN-F, even though the protein was expressed on the cell surface. A construct in which the PI3-HN cytoplasmic tail and transmembrane domain were substituted for those of SN in the SN(140) chimera still maintained the ability to promote cell fusion. These results indicate that a region including only 82 amino acids in the extracellular domain, adjacent to the transmembrane domain of the SN-HN protein, is important for interaction with the SN-F protein and promotion of cell fusion.  相似文献   

12.
The glycoprotein complex of paramyxoviruses mediates receptor binding and membrane fusion. In particular, the measles virus (MV) fusion (F) protein executes membrane fusion, after receptor binding by the hemagglutinin (H) protein. Structures and single amino acids influencing fusion function have been identified in the F-protein ectodomain and cytoplasmic tail, but not in its transmembrane (TM) region. Since this region influences function of the envelope proteins of other viruses, we examined its role in the MV F protein. Alanine-scanning mutagenesis revealed that an F protein with a single mutation of a central TM region leucine (L507A) was more fusogenic than the unmodified F protein while retaining similar kinetics of proteolytic processing. In contrast, substitution of residues located near the edges of the lipid bilayer reduced fusion activity. This was true not only when the mutated F proteins were coexpressed with H but also in the context of infections with recombinant viruses. Analysis of the H-F complexes with reduced fusion activities revealed that more precursor (F0) than activated (F1+2) protein coprecipitated with H. In contrast, in complexes with enhanced fusion activity, including H-FL507A, the F0/F1+2 ratio shifted toward F1+2. Thus, fusion activity correlated with an active F-H protein complex, and the MV F protein TM region modulated availability of this complex.  相似文献   

13.
Purified plasma membranes attached to polycationic polyacrylamide beads by their external surface were isolated from BHK cells infected with Sendai virus. Each of the viral proteins could be identified in the membranes of infected cells. Proteolysis with trypsin, which digests only the cytoplasmic surface of these membranes (because the external surface is protected by its attachment to beads), revealed that the internal proteins, L, P, NP, and M, were present on the cytoplasmic surface of the membrane and that small segments of the viral envelope glycoproteins, HN and F0, were partially exposed on the cytoplasmic surface. Since the major portions of HN and F0 are known to be present on the external membrane surface, these glycoproteins are transmembrane proteins before Sendai virus budding in infected cells.  相似文献   

14.
We sought to develop a live attenuated parainfluenza virus type 2 (PIV2) vaccine strain for use in infants and young children, using reverse genetic techniques that previously were used to rapidly produce a live attenuated PIV1 vaccine candidate. The PIV1 vaccine candidate, designated rPIV3-1cp45, was generated by substituting the full-length HN and F proteins of PIV1 for those of PIV3 in the attenuated cp45 PIV3 vaccine candidate (T. Tao et al., J. Virol. 72:2955-2961, 1998; M. H. Skiadopoulos et al., Vaccine 18:503-510, 1999). However, using the same strategy, we failed to recover recombinant chimeric PIV3-PIV2 isolate carrying the full-length PIV2 glycoproteins in a wild-type PIV3 backbone. Viable PIV3-PIV2 chimeras were recovered when chimeric HN and F open reading frames (ORFs) rather than complete PIV2 F and HN ORFs were used to construct the full-length cDNA. The recovered viruses, designated rPIV3-2CT, in which the PIV2 ectodomain and transmembrane domain were fused to the PIV3 cytoplasmic domain, and rPIV3-2TM, in which the PIV2 ectodomain was fused to the PIV3 transmembrane and cytoplasmic tail domain, possessed similar in vitro and in vivo phenotypes. Thus, it appeared that only the cytoplasmic tail of the HN or F glycoprotein of PIV3 was required for successful recovery of PIV3-PIV2 chimeras. Although rPIV3-2CT and rPIV3-2TM replicated efficiently in vitro, they were moderately to highly attenuated for replication in the respiratory tracts of hamsters, African green monkeys (AGMs), and chimpanzees. This unexpected finding indicated that chimerization of the HN and F proteins of PIV2 and PIV3 itself specified an attenuation phenotype in vivo. Despite this attenuation, these viruses were highly immunogenic and protective against challenge with wild-type PIV2 in hamsters and AGMs, and they represent promising candidates for clinical evaluation as a vaccine against PIV2. These chimeric viruses were further attenuated by the addition of 12 mutations of PIV3cp45 which lie outside of the HN and F genes. The attenuating effects of these mutations were additive with that of the chimerization, and thus inclusion of all or some of the cp45 mutations provides a means to further attenuate the PIV3-PIV2 chimeric vaccine candidates if necessary.  相似文献   

15.
Paramyxoviruses enter host cells by fusing the viral envelope with a host cell membrane. Fusion is mediated by the viral fusion (F) protein, and it undergoes large irreversible conformational changes to cause membrane merger. The C terminus of PIV5 F contains a membrane-proximal 7-residue external region (MPER), followed by the transmembrane (TM) domain and a 20-residue cytoplasmic tail. To study the sequence requirements of the F protein C terminus for fusion, we constructed chimeras containing the ectodomain of parainfluenza virus 5 F (PIV5 F) and either the MPER, the TM domain, or the cytoplasmic tail of the F proteins of the paramyxoviruses measles virus, mumps virus, Newcastle disease virus, human parainfluenza virus 3, and Nipah virus. The chimeras were expressed, and their ability to cause cell fusion was analyzed. The chimeric proteins were variably expressed at the cell surface. We found that chimeras containing the ectodomain of PIV5 F with the C terminus of other paramyxoviruses were unable to cause cell fusion. Fusion could be restored by decreasing the activation energy of refolding through introduction of a destabilizing mutation (S443P). Replacing individual regions, singly or doubly, in the chimeras with native PIV5 F sequences restored fusion to various degrees, but it did not have an additive effect in restoring activity. Thus, the F protein C terminus may be a specific structure that only functions with its cognate ectodomain. Alanine scanning mutagenesis of MPER indicates that it has a regulatory role in fusion since both hyperfusogenic and hypofusogenic mutations were found.  相似文献   

16.
[3H]palmitic acid was metabolically incorporated into the viral fusion protein (F) of Edmonston or freshly isolated measles virus (MV) during infection of human lymphoid or Vero cells. The uncleaved precursor F0 and the F1 subunit from infected cells and extracellular virus were both labeled, indicating that palmitoylation can take place prior to F0 cleavage and that palmitoylated F protein was incorporated into virus particles. [3H]palmitic acid was released from F protein upon hydroxylamine or dithiothreitol treatment, indicating a thioester linkage. In cells transfected with the cloned MV F gene, in which the cysteines located in the intracytoplasmic and transmembrane domains (Cys 506, 518, 519, 520, and 524) were replaced by serine, a major reduction of [3H]palmitic acid incorporation was observed for F mutated at Cys 506 and, to a lesser extent, at Cys 518 and Cys 524. We also observed incorporation of [3H]palmitic acid in the F1 subunit of canine distemper virus F protein. Cell fusion induced by cotransfection of cells with MV F and H (hemagglutinin) genes was significantly reduced after replacement of Cys 506 or Cys 519 with serine in the MV F gene. Transfection with the F gene with a mutation for Cys 518 abolished cell fusion, although less mutant protein was detected on the cell surface. These results suggest that the F protein transmembrane domain cysteines 506 and 518 participate in structures involved in cell fusion, possibly mediated by palmitoylation.  相似文献   

17.
Moll M  Klenk HD  Maisner A 《Journal of virology》2002,76(14):7174-7186
The generation of replication-competent measles virus (MV) depends on the incorporation of biologically active, fusogenic glycoprotein complexes, which are required for attachment and penetration into susceptible host cells and for direct virus spread by cell-to-cell fusion. Whereas multiple studies have analyzed the importance of the ectodomains of the MV glycoproteins hemagglutinin (H) and fusion protein (F), we have investigated the role of the cytoplasmic tails of the F and H proteins for the formation of fusogenic complexes. Deletions in the cytoplasmic tails of transiently expressed MV glycoproteins were found to have varying effects on receptor binding, fusion, or fusion promotion activity. F tail truncation to only three amino acids did not affect fusion capacity. In contrast, truncation of the H cytoplasmic tail was limited. H protein mutants with cytoplasmic tails of <14 residues no longer supported F-mediated cell fusion, predominantly due to a decrease in surface expression and receptor binding. This indicates that a minimal length of the H protein tail of 14 amino acids is required to ensure a threshold local density to have sufficient accumulation of fusogenic H-F complexes. By using reverse genetics, a recombinant MV with an F tail of three amino acids (rMV-FcDelta30), as well as an MV with an H tail of 14 residues (rMV-HcDelta20), could be rescued, whereas generation of viruses with shorter H tails failed. Thus, glycoprotein truncation does not interfere with the successful generation of recombinant MV if fusion competence is maintained.  相似文献   

18.
To determine the role of amino acid sequences of the hemagglutinin-neuraminidase (HN) cytoplasmic tail in Newcastle disease virus (NDV) replication and pathogenicity, we generated recombinant NDVs with a deletion or point mutation in the N-terminal cytoplasmic tail. The first 2-amino-acid deletion in the cytoplasmic tail did not affect the biological characteristics of NDV. However, a 4-amino-acid deletion and the substitution of alanine for serine at position 6 affected cell fusion, pathogenicity, and colocalization of the HN and M proteins of NDV, indicating that these residues of the HN cytoplasmic tail are critical for its specific incorporation into virions.Newcastle disease virus (NDV) causes a highly contagious respiratory and neurologic disease in chickens, leading to severe economic losses in the poultry industry worldwide (1). NDV is a member of the family Paramyxoviridae and has a nonsegmented, negative-sense RNA genome consisting of six genes (3′-NP-P-M-F-HN-L-5′) (7). Infection of host cells by NDV is accomplished through the interaction of two surface glycoproteins, the fusion (F) and hemagglutinin-neuraminidase (HN) proteins. The F protein directs the membrane fusion between the viral and cellular membranes, while the HN protein mediates attachment to sialic acid, has neuraminidase activity, and plays a role in fusion promotion (4).The HN protein of NDV is a type II transmembrane glycoprotein and possesses three spatially distinct domains: the ectodomain, transmembrane domain, and cytoplasmic tail. The globular ectodomain contains the sites for receptor binding and neuraminidase activity, and the transmembrane domain anchors to viral envelopes (8). The cytoplasmic tail domain contains 26 highly conserved amino acids whose functions are not well-known. In a plasmid-based expression system, truncation (23 amino acids) of the cytoplasmic tail caused improper orientation of the HN protein in the membrane insertion (13). In other paramyxoviruses, cytoplasmic tails of the HN proteins are known to play crucial roles in virus budding and assembly (10, 12). Our unsuccessful attempt to recover a recombinant NDV (rNDV) with complete deletion of the HN cytoplasmic tail also suggested that the cytoplasmic tail is required for assembly and budding of NDV. Therefore, in this study, we determined the role of amino acid sequences of the cytoplasmic tail in the NDV replication cycle. Since essential regions of the HN cytoplasmic tail for virus replication are unknown, we consecutively deleted the first 6 nucleotides (nt), 12 nt, or 18 nt of the HN cytoplasmic tail in a full-length antigenomic cDNA of NDV intermediate virulent (mesogenic) strain Beaudette C (BC) (6), thus maintaining the “rule of six” for the NDV genome (Fig. (Fig.1A).1A). rNDVs were recovered using our standard protocol (6). We recovered rNDVs containing 2-amino-acid deletion and 4-amino-acid deletion of the HN cytoplasmic tail (rBC/HNΔ2 and rBC/HNΔ4, respectively), indicating that only these 4 amino acids are dispensable in generating infectious virions. Since rNDV containing 6-amino-acid deletion of the HN cytoplasmic tail could not be recovered, we wanted to know the role of amino acids at positions 5 and 6 in NDV replications. The serine residue at position 6 is a potential phosphorylation site. Therefore, to determine whether phosphorylation at this site is crucial for recovery of NDV, we additionally generated rNDVs with substitution of alanine and glutamic acid for serine (rBC/HNS6A and rBC/HNS6E, respectively) to confirm its crucial role in the recovery of rNDV.Open in a separate windowFIG. 1.Constructs of recombinant NDVs containing a deletion or point mutation in the N-terminal cytoplasmic tail of the HN protein and replication and fusion index of recovered viruses in infected cells. (A) Consecutively, 6 nt, 12 nt, or 18 nt of mRNA of the HN cytoplasmic tail in a full-length antigenomic cDNA of NDV was deleted. Deletions in the HN cytoplasmic tails are indicated by the large boldface dashes. In addition, serine at position 6 was substituted with alanine and glutamic acid was substituted by changing guanine to cytidine and adenosine, respectively. (B) In vitro replication of the mutant viruses was determined in virus-infected DF-1 cells at an MOI of 0.01. The viral titers were determined by plaque assay. (C) The fusion index was determined in virus-infected Vero cells at an MOI of 0.1. Cells were stained with hematoxylin-eosin, and the fusion index was calculated as a mean number of nuclei per cell. The assay was performed three times.In vitro replication of recovered viruses was determined by plaque assay in virus-infected DF-1 cells at a multiplicity of infection (MOI) of 0.01 (5). All mutant viruses and the parental virus, rBC, grew to similar titers, indicating that alteration of the HN cytoplasmic tails did not affect their in vitro replication (Fig. (Fig.1B).1B). Although the rBC/HNΔ4 mutant had grown well up to 24 h postinfection, a reduction of the viral titer was detected thereafter with rapid and extensive induction of syncytia. Therefore, we determined fusion promotion activity of the mutant viruses by quantitating syncytia in virus-infected Vero cells at an MOI of 0.1 at 30 h postinfection (8) and confirmed increased fusion promotion activity of rBC/HNΔ4 followed by rBC/HNS6A compared to that of rBC (Fig. (Fig.1C).1C). Similarly, enhanced fusion activity was observed in other cytoplasmic tail-truncated paramyxoviruses, such as simian virus 5 and measles virus (2, 9). It has been postulated that interaction of matrix (M) protein with the cytoplasmic tails of the glycoproteins involves in a fusion-refractory conformation at the early stage of viral maturation (2). Therefore, these altered HN cytoplasmic tails could assist NDV in gaining its cell fusion competence by modulating this fusion-refractory conformation.In general, the levels of the HN protein contents on the surfaces of virus-infected cells and in the virus particles were more affected by point mutation of serine than by truncation of the cytoplasmic tail. We analyzed surface expression of the HN protein on virus-infected DF-1 cells at an MOI of 0.1. At 24 h postinfection, the cells were labeled with a monoclonal antibody against the NDV HN protein followed by anti-Alexa Fluor 488 conjugate, fixed with 4% paraformaldehyde, and analyzed by a fluorescence-activated cell sorter (AriaII; BD Bioscience) with Flowjo program (Tree Star, Inc.) (Fig. (Fig.2A).2A). The percentages of cells expressing the HN proteins were 89 (rBC), 78 (rBC/HNΔ2), 71 (rBC/HNΔ4), 64 (rBC/HNS6A), and 53 (rBC/HNS6E). To analyze incorporation of the HN proteins into the viral particles, the parental and mutant viruses harvested from allantoic fluid samples were purified through a 30% sucrose cushion. The viral proteins were separated on an 8% sodium dodecyl sulfate-polyacrylamide gel (Fig. (Fig.2B).2B). We first examined whether the mutant viruses incorporated the same levels of other viral proteins. This assay was performed by determining the ratios of the P protein to M protein. We found that similar levels of the P and M proteins were present among the different mutant viruses (Fig. (Fig.2B).2B). We then measured the levels of the HN proteins incorporated into the virus particles by determining the ratios of the HN protein to M protein (Fig. (Fig.2C).2C). The pattern of incorporation of the HN proteins into the virus particles was similar with their cell surface expression. The HN protein contents of rBC/HNΔ2 and rBC/HNΔ4 were not significantly different from that of the parental virus (P > 0.05), indicating that truncation of the cytoplasmic tail did not impair its incorporation into the viral particles. In contrast, substitution of glutamic acid for serine decreased incorporation of the HN protein into the viral particles, indicating that serine plays an important role in both cell surface expression of the HN protein and its incorporation into the viral particles.Open in a separate windowFIG. 2.Effect of alteration of the HN cytoplasmic tail on incorporation of the HN proteins into viral particles and their surface expression in DF-1 cells. (A) Surface expression of the NDV HN protein in DF-1 cells was analyzed by a fluorescence-activated cell sorter. At 24 h postinfection, DF-1 cells infected with each virus were stained with monoclonal antibody against the HN protein followed by anti-Alexa Fluor 488 conjugate. (B) Ultracentrifuge-purified viruses from infected allantoic fluid samples were separated by electrophoresis, and the gel was then stained with Coomassie brilliant blue. (C) Ratios of HN protein to M-protein levels from the parental virus and the HN cytoplasmic tail mutant viruses were quantified.We further determined the effect of cytoplasmic tail alteration on the pathogenicity of NDV in embryonated eggs and chicks (Table (Table1).1). The mean death time (MDT) was determined as the mean time (h) for the minimum lethal dose of virus to kill all the embryos after inoculation of 9-day-old specific-pathogen-free (SPF) embryonated chicken eggs with virus (1). The criteria for classifying the virulence of NDV strains are as follows: virulent strains take <60 h to kill embryos, intermediate virulent strains take 60 to 90 h to kill embryos, and avirulent strains take >90 h to kill embryos. Two mutant viruses (rBC/HNΔ2 and rBC/HNS6E) showed similar values of MDT compared to rBC (59 h). In contrast, the MDTs of rBC/HNΔ4 and rBC/HNS6A were 50 h and 51 h, respectively. Increased pathogenicity of these two mutants was also confirmed by an intracerebral pathogenicity index (ICPI) test in 1-day-old SPF chicks (1). The scale of the ICPI value in evaluating the virulence of NDV strains is from 0.00 (avirulent strains) to 2.00 (highly virulent NDV strains). The rBC/HNΔ4 virus had the highest ICPI value (1.61), followed by rBC/HNS6A (ICPI value of 1.58), among the parental and mutant viruses, probably due to their enhanced fusion promotion activity. In contrast, rBC/HNS6E had the lowest ICPI value (1.41), which would be associated with decreased HN protein contents detected in the viral particles and virus-infected cells. In our previous study, decreased HN protein contents in virus particles due to complete deletion of 5′ untranslated regions of the HN gene also resulted in attenuation of the virus in chickens (14). Consistently, rBC/HNΔ2 showed biological characteristics and pathogenicity similar to those of the parental virus, suggesting that aspartic acid and arginine are indispensable for the HN cytoplasmic tail of NDV.

TABLE 1.

Pathogenicity of the HN cytoplasmic tail mutant viruses in embryonated eggs and chicks
VirusMDT (h)aICPIb
rBC581.49
rBC/HNΔ2591.51
rBC/HNΔ4501.61
rBC/HNS6A511.58
rBC/HNS6E621.41
Open in a separate windowaThe mean time (in hours) for the minimum lethal dose of virus to kill all the inoculated embryos. NDV strains were classified by the following criteria: virulent strains take <60 h to kill embryos, intermediate virulent strains take 60 to 90 h to kill embryos, and avirulent strains take >90 h to kill embryos.bPathogenicity of NDV in 1-day-old SPF chicks was evaluated by the ICPI value: virulent strains had ICPI values of 1.5 to 2.0, intermediate virulent strains had ICPI values of 1.0 to 1.5, and avirulent strains had ICPI values of 0.0 to 0.5.The M protein plays a major role in virus assembly through its interaction with envelope glycoproteins and with the membranes of infected cells (11). To gain insight into the function of the amino acid sequences of the HN cytoplasmic tail in virus assembly, colocalization of the HN and M proteins was determined by confocal microscopy (LSM 510; Zeiss). Detection of the M and HN proteins was facilitated by coexpressing M protein and each altered HN protein using the pCAGGS expression system in 293T cells. In particular, the open reading frame of the M gene had been fused with an influenza virus hemagglutinin epitope tag (7 amino acid residues) followed by a stop codon and cloned into pCAGGS. After 24 h of transfection, the cells were fixed, permeabilized, stained with a monoclonal antibody against the NDV HN protein followed by anti-Alexa Fluor 488 and anti-HA Alexa Fluor 594 conjugates, and analyzed by confocal microscopy. The M and wild-type HN proteins were distributed in the nucleus and cytoplasm and in the cytoplasm, respectively, leading to their colocalization in the cytoplasm of infected cells (Fig. (Fig.3A).3A). In contrast, cytoplasmic tail-altered HN proteins (4-amino-acid deletion and substitution of alanine for serine) were dominantly found on the cell surface with their colocalization with the M protein, indicating reduction of specificity in membrane insertion of these HN proteins (Fig. 3B and C). Furthermore, no colocalization of the 6-amino-acid deletion of cytoplasmic tail-altered HN protein with the M protein was detected (Fig. (Fig.3D),3D), suggesting that this alteration had affected incorporation of the HN protein into virus particles and consequently virus recovery. Other paramyxoviruses (e.g., simian virus 5 and human respiratory syncytial virus) also showed a loss of intracellular interaction between the M protein and glycoproteins containing cytoplasmic tail-truncated domains (3, 12).Open in a separate windowFIG. 3.Localization of the NDV HN and M proteins in 293T cells. The M protein fused with an influenza virus hemagglutinin epitope tag and each HN variant containing altered cytoplasmic tails were expressed using the pCAGGS expression system in 293T cells. The cells were fixed with 4% paraformaldehyde, permeabilized with 0.2% Triton X-100, stained with a monoclonal antibody against the NDV HN protein followed by anti-Alexa Fluor 488 (green; HN) and anti-HA Alexa Fluor 594 (red; M) conjugates and analyzed by laser-scanning microscopy. (A) Wild-type HN, (B) HN Δ4, (C) HN S6A, and (D) HN Δ6.In summary, we demonstrate that the cytoplasmic tail of HN plays a crucial role in the NDV life cycle. Our data suggest that the first 2 amino acids of the cytoplasmic tail are not absolutely required for NDV replication, but amino acids at positions 4 through 6 are critical for specific insertion of the HN protein into virion particles. Furthermore, our results indicate that the cytoplasmic tail of HN protein modulates the fusion activity of NDV. It will also be necessary to determine whether alteration of the HN cytoplasmic tail can affect interaction of the HN protein with the F protein.  相似文献   

19.
A mutational analysis was used to identify structural domains that are important for exocytic transport and proteolytic cleavage of the mouse mammary tumor virus (MMTV) glycoprotein, which is expressed as a multidomain polyprotein. Rat HTC hepatoma cells were transfected with the MMTV glycoprotein gene driven by the constitutive Rous sarcoma virus promoter, with mutant genes encoding a series of polypeptide truncations or with a defective MMTV provirus containing a premature termination codon in the viral glycoprotein gene. Efficient proteolytic maturation and transport of MMTV glycoproteins to the cell surface or extracellular environment required the presence of the transmembrane domain but not the cytoplasmic tail. Two stable truncations retaining the hydrophobic region of the ectodomain in the absence of the transmembrane domain and cytoplasmic tail (trgp67 and trgp58) remained in endoglycosidase H sensitive and uncleaved forms. One of these truncations, trgp58, appeared to be tightly associated with intracellular membranes and strongly bound by heavy chain binding protein, whereas the other truncation, trgp67, was a soluble component of the lumen and persists intracellularly by a heavy chain binding protein-independent pathway. The truncated MMTV glycoprotein additionally lacking the hydrophobic region of the ectodomain was efficiently secreted. Taken together, our results demonstrate that the hydrophobic transmembrane domain of the MMTV glycoprotein is required for proper transport and proteolytic processing, whereas, in the absence of the transmembrane domain, the presence of a hydrophobic region of the ectodomain correlated with retention at an early step in the exocytic pathway.  相似文献   

20.
The role of the sequence of transmembrane and cytoplasmic/intraviral domains of influenza virus hemagglutinin (HA, subtype H7) for HA-mediated membrane fusion was explored. To analyze the influence of the two domains on the fusogenic properties of HA, we designed HA-chimeras in which the cytoplasmic tail and/or transmembrane domain of HA was replaced with the corresponding domains of the fusogenic glycoprotein F of Sendai virus. These chimeras, as well as constructs of HA in which the cytoplasmic tail was replaced by peptides of human neurofibromin type1 (NF1) or c-Raf-1, NF78 (residues 1441 to 1518), and Raf81 (residues 51 to 131), respectively, were expressed in CV-1 cells by using the vaccinia virus-T7 polymerase transient-expression system. Wild-type and chimeric HA were cleaved properly into two subunits and expressed as trimers. Membrane fusion between CV-1 cells and bound human erythrocytes (RBCs) mediated by parental or chimeric HA proteins was studied by a lipid-mixing assay with the lipid-like fluorophore octadecyl rhodamine B chloride (R18). No profound differences in either extent or kinetics could be observed. After the pH was lowered, the above proteins also induced a flow of the aqueous fluorophore calcein from preloaded RBCs into the cytoplasm of the protein-expressing CV-1 cells, indicating that membrane fusion involves both leaflets of the lipid bilayers and leads to formation of an aqueous fusion pore. We conclude that neither HA-specific sequences in the transmembrane and cytoplasmic domains nor their length is crucial for HA-induced membrane fusion activity.  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