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 31 毫秒
1.
Canine parvovirus (CPV) and feline panleukopenia virus (FPV) differ in their ability to infect dogs and dog cells. Canine cell infection is a specific property of CPV and depends on the ability of the virus to bind the canine transferrin receptor (TfR), as well as other unidentified factors. Three regions in the capsid structure, located around VP2 residues 93, 300, and 323, can all influence canine TfR binding and canine cell infection. These regions were compared in the CPV and FPV capsid structures that have been determined, as well as in two new structures of CPV capsids that contain substitutions of the VP2 Asn-93 to Asp and Arg, respectively. The new structures, determined by X-ray crystallography to 3.2 and 3.3 A resolutions, respectively, clearly showed differences in the interactions of residue 93 with an adjacent loop on the capsid surface. Each of the three regions show small differences in structure, but each appears to be structurally independent of the others, and the changes likely act together to affect the ability of the capsid to bind the canine TfR and to infect canine cells. This emphasizes the complex nature of capsid alterations that change the virus-cell interaction to allow infection of cells from different hosts.  相似文献   

2.
Canine parvovirus (CPV) and feline panleukopenia virus (FPV) capsids bind to the transferrin receptors (TfRs) of their hosts and use these receptors to infect cells. The binding is partially host specific, as FPV binds only to the feline TfR, while CPV binds to both the canine and feline TfRs. The host-specific binding is controlled by a combination of residues within a raised region of the capsid. To define the TfR structures that interact with the virus, we altered the apical domain of the feline or canine TfR or prepared chimeras of these receptors and tested the altered receptors for binding to FPV or CPV capsids. Most changes in the apical domain of the feline TfR did not affect binding, but replacing Leu221 with Ser or Asp prevented receptor binding to either FPV or CPV capsids, while replacing Leu221 with Lys resulted in a receptor that bound only to CPV but not to FPV. Analysis of recombinants of the feline and canine TfRs showed that sequences controlling CPV-specific binding were within the apical domain and that more than one difference between these receptors determined the CPV-specific binding of the canine TfR. Single changes within the canine TfR which removed a single amino acid insertion or which eliminated a glycosylation site gave that receptor the expanded ability to bind to FPV and CPV. In some cases, binding of capsids to mutant receptors did not result in infection, suggesting a structural role for the receptor in cell infection by the viruses.  相似文献   

3.
The cell infection processes and host ranges of canine parvovirus (CPV) and feline panleukopenia virus (FPV) are controlled by their capsid interactions with the transferrin receptors (TfR) on their host cells. Here, we expressed the ectodomains of wild-type and mutant TfR and tested those for binding to purified viral capsids and showed that different naturally variant strains of the viruses were associated with variant interactions with the receptors which likely reflect the optimization of the viral infection processes in the different hosts. While all viruses bound the feline TfR, reflecting their tissue culture host ranges, a naturally variant mutant of CPV (represented by the CPV type-2b strain) that became the dominant virus worldwide in 1979 showed significantly lower levels of binding to the feline TfR. The canine TfR ectodomain did not bind to a detectable level in the in vitro assays, but this appears to reflect the naturally low affinity of that interaction, as only low levels of binding were seen when the receptor was expressed on mammalian cells; however, that was sufficient to allow endocytosis and infection. The apical domain of the canine TfR controls the specific interaction with CPV capsids, as a canine TfR mutant altering a glycosylation site in that domain bound FPV, CPV-2, and CPV-2b capsids efficiently. Enzymatic removal of the N-linked glycans did not allow FPV binding to the canine TfR, suggesting that the protein sequence difference is itself important. The purified feline TfR inhibited FPV and CPV-2 binding and infection of feline cells but not CPV-2b, indicating that the receptor binding may be able to prevent the attachment to the same receptor on cells.  相似文献   

4.
S F Chang  J Y Sgro    C R Parrish 《Journal of virology》1992,66(12):6858-6867
Canine parvovirus (CPV) and feline panleukopenia virus (FPV) are over 98% similar in DNA sequence but have specific host range, antigenic, and hemagglutination (HA) properties which were located within the capsid protein gene. In vitro mutagenesis and recombination were used to prepare 16 different recombinant genomic clones, and viruses derived from those clones were analyzed for their in vitro host range, antigenic, and HA properties. The region of CPV from 59 to 91 map units determined the ability to replicate in canine cells. A complex series of interactions was observed among the individual sequence differences between 59 and 73 map units. The canine host range required that VP2 amino acids (aa) 93 and 323 both be the CPV sequence, and those two CPV sequences introduced alone into FPV greatly increased viral replication in canine cells. Changing any one of aa 93, 103, or 323 of CPV to the FPV sequence either greatly decreased replication in canine cells or resulted in an inviable plasmid. The Asn-Lys difference of aa 93 alone was responsible for the CPV-specific epitope recognized by monoclonal antibodies. An FPV-specific epitope was affected by aa 323. Amino acids 323 and 375 together determined the pH dependence of HA. Amino acids involved in the various specific properties were all around the threefold spikes of the viral particle.  相似文献   

5.
Canine parvovirus (CPV) is a host range variant of a feline virus that acquired the ability to infect dogs through changes in its capsid protein. Canine and feline viruses both use the feline transferrin receptor (TfR) to infect feline cells, and here we show that CPV infects canine cells through its ability to specifically bind the canine TfR. Receptor binding on host cells at 37 degrees C only partially correlated with the host ranges of the viruses, and an intermediate virus strain (CPV type 2) bound to higher levels on cells than did either the feline panleukopenia virus or a later strain of CPV. During the process of adaptation to dogs the later variant strain of CPV gained the ability to more efficiently use the canine TfR for infection and also showed reduced binding to feline and canine cells compared to CPV type 2. Differences on the top and the side of the threefold spike of the capsid surface controlled specific TfR binding and the efficiency of binding to feline and canine cells, and these differences also determined the cell infection properties of the viruses.  相似文献   

6.
Canine parvovirus (CPV) enters and infects cells by a dynamin-dependent, clathrin-mediated endocytic pathway, and viral capsids colocalize with transferrin in perinuclear vesicles of cells shortly after entry (J. S. L. Parker and C. R. Parrish, J. Virol. 74:1919-1930, 2000). Here we report that CPV and feline panleukopenia virus (FPV), a closely related parvovirus, bind to the human and feline transferrin receptors (TfRs) and use these receptors to enter and infect cells. Capsids did not detectably bind or enter quail QT35 cells or a Chinese hamster ovary (CHO) cell-derived cell line that lacks any TfR (TRVb cells). However, capsids bound and were endocytosed into QT35 cells and CHO-derived TRVb-1 cells that expressed the human TfR. TRVb-1 cells or TRVb cells transiently expressing the feline TfR were susceptible to infection by CPV and FPV, but the parental TRVb cells were not. We screened a panel of feline-mouse hybrid cells for susceptibility to FPV infection and found that only those cells that possessed feline chromosome C2 were susceptible. The feline TfR gene (TRFC) also mapped to feline chromosome C2. These data indicate that cell susceptibility for these viruses is determined by the TfR.  相似文献   

7.
Canine parvovirus (CPV) emerged in 1978 as a host range variant of feline panleukopenia virus (FPV). This change of host was mediated by the mutation of five residues on the surface of the capsid. CPV and FPV enter cells by endocytosis and can be taken up by many non-permissive cell lines, showing that their host range and tissue specificity are largely determined by events occurring after cell entry.We have determined the structures of a variety of strains of CPV and FPV at various pH values and in the presence or absence of Ca(2+). The largest structural difference was found to occur in a flexible surface loop, consisting of residues 359 to 375 of the capsid protein. This loop binds a divalent calcium ion in FPV and is adjacent to a double Ca(2+)-binding site, both in CPV and FPV. Residues within the loop and those associated with the double Ca(2+)-binding site were found to be essential for virus infectivity. The residues involved in the double Ca(2+)-binding site are conserved only in FPV and CPV.Our results show that the loop conformation and the associated Ca(2+)-binding are influenced by the Ca(2+) concentration, as well as pH. These changes are correlated with the ability of the virus to hemagglutinate erythrocytes. The co-localization of hemagglutinating activity and host range determinants on the virus surface implies that these properties may be functionally linked. We speculate that the flexible loop and surrounding regions are involved in binding an as yet unidentified host molecule and that this interaction influences host range.  相似文献   

8.
Canine parvovirus (CPV) emerged as an apparently new virus during the mid-1970s. The origin of CPV is unknown, but a variation from feline panleukopenia virus (FPV) or another closely related parvovirus is suspected. Here we examine the in vitro and in vivo canine and feline host ranges of CPV and FPV. Examination of three canine and six feline cell lines and mitogen-stimulated canine and feline peripheral blood lymphocytes revealed that CPV replicates in both canine and feline cells, whereas FPV replicates efficiently only in feline cells. The in vivo host ranges were unexpectedly complex and distinct from the in vitro host ranges. Inoculation of dogs with FPV revealed efficient replication in the thymus and, to some degree, in the bone marrow, as shown by virus isolation, viral DNA recovery, and Southern blotting and by strand-specific in situ hybridization. FPV replication could not be demonstrated in mesenteric lymph nodes or in the small intestine, which are important target tissues in CPV infection. Although CPV replicated well in all the feline cells tested in vitro, it did not replicate in any tissue of cats after intramuscular or intravenous inoculation. These results indicate that these viruses have complex and overlapping host ranges and that distinct tissue tropisms exist in the homologous and heterologous hosts.  相似文献   

9.
The unique N-terminal region of the parvovirus VP1 capsid protein is required for infectivity by the capsids but is not required for capsid assembly. The VP1 N terminus contains a number of groups of basic amino acids which resemble classical nuclear localization sequences, including a conserved sequence near the N terminus comprised of four basic amino acids, which in a peptide can act to transport other proteins into the cell nucleus. Testing with a monoclonal antibody recognizing residues 2 to 13 of VP1 (anti-VP1-2-13) and with a rabbit polyclonal serum against the entire VP1 unique region showed that the VP1 unique region was not exposed on purified capsids but that it became exposed after treatment of the capsids with heat (55 to 75 degrees C), or urea (3 to 5 M). A high concentration of anti-VP1-2-13 neutralized canine parvovirus (CPV) when it was incubated with the virus prior to inoculation of cells. Both antibodies blocked infection when injected into cells prior to virus inoculation, but neither prevented infection by coinjected infectious plasmid DNA. The VP1 unique region could be detected 4 and 8 h after the virus capsids were injected into cells, and that sequence exposure appeared to be correlated with nuclear transport of the capsids. To examine the role of the VP1 N terminus in infection, we altered that sequence in CPV, and some of those changes made the capsids inefficient at cell infection.  相似文献   

10.
The feline and canine transferrin receptors (TfRs) bind canine parvovirus to host cells and mediate rapid capsid uptake and infection. The TfR and its ligand transferrin have well-described pathways of endocytosis and recycling. Here we tested several receptor-dependent steps in infection for their role in virus infection of cells. Deletions of cytoplasmic sequences or mutations of the Tyr-Thr-Arg-Phe internalization motif reduced the rate of receptor uptake from the cell surface, while polar residues introduced into the transmembrane sequence resulted in increased degradation of transferrin. However, the mutant receptors still mediated efficient virus infection. In contrast, replacing the cytoplasmic and transmembrane sequences of the feline TfR with those of the influenza virus neuraminidase (NA) resulted in a receptor that bound and endocytosed the capsid but did not mediate viral infection. This chimeric receptor became localized to detergent-insoluble membrane domains. To test the effect of structural virus receptor interaction on infection, two chimeric receptors were prepared which contained antibody-variable domains that bound the capsid in place of the TfR ectodomain. These chimeric receptors bound CPV capsids and mediated uptake but did not result in cell infection. Adding soluble feline TfR ectodomain to the virus during that uptake did not allow infection.  相似文献   

11.
Canine parvovirus (CPV) and feline panleukopenia virus (FPV) are closely related parvoviruses that differ in their host ranges for cats and dogs. Both viruses bind their host transferrin receptor (TfR), enter cells by clathrin-mediated endocytosis, and traffic with that receptor through endosomal pathways. Infection by these viruses appears to be inefficient and slow, with low numbers of virions infecting the cell after a number of hours. Species-specific binding to TfR controls viral host range, and in this study FPV and strains of CPV differed in the levels of cell attachment, uptake, and infection in canine and feline cells. During infection, CPV particles initially bound and trafficked passively on the filopodia of canine cells while they bound to the cell body of feline cells. That binding was associated with the TfR as it was disrupted by anti-TfR antibodies. Capsids were taken up from the cell surface with different kinetics in canine and feline cells but, unlike transferrin, most did not recycle. Capsids labeled with fluorescent markers were seen in Rab5-, Rab7-, or Rab11-positive endosomal compartments within minutes of uptake, but reached the nucleus. Constitutively active or dominant negative Rab mutants changed the intracellular distribution of capsids and affected the infectivity of virus in cells.Cell infection by animal viruses involves a specific sequence of steps that deliver the virus and its genome from the cell surface to the compartment where replication can occur. For nonenveloped viruses, infection initiates with binding to a specific cell receptor and uptake into the cell by receptor-mediated endocytosis. Various factors can control the process of viral uptake, including the characteristics of the receptor(s) bound by the virus and its signaling and endocytic properties, the affinity of the virus for the receptor, and the structural features of the interaction in different environments (36, 61). Receptors may be located on the cell body or may also be displayed on the extended lamellipodia or filopodia with greater surface areas. Viruses binding to filopodia can be either passively delivered to the cell body for endocytosis by dynamic movement of the entire structure or actively trafficked by retrograde actin transport as well as the action of myosin-2 motors on the actin (32, 57). Cross-linking and clustering of receptors by viral particles can influence the rate and pathways of uptake from the cell surface (23), and many viral receptors activate signaling pathways that alter the structure of the underlying cytoskeleton to enhance uptake (see, e.g., references 12, 30, and 51). Receptor-bound viruses then enter one or more endosomal pathways that results in the capsid being enclosed in vesicles and trafficked within the endosomal pathways of the cell, where clustered virus and receptors (23) may undergo structural alterations upon exposure to conditions such as low pH or proteases (36, 61). The specific receptor-mediated binding and entry pathways often provide signals for viruses that allow endosomal escape and establish infection. A variety of markers of the endosomal compartments have been used in studies of viral entry. Rab proteins are monomeric small GTPases which regulate endosomal membrane trafficking, and specific Rab proteins are associated with different endosomal compartments. Among the many Rab proteins in the cells, Rab5 is primarily associated with the early endosome and regulates trafficking through that compartment, Rab7 is associated with the late endosome, and Rab11 is associated with the recycling endosome (14, 58). Tracking viral particles within the endosomal pathways during cell entry has been used to define the steps in the entry and infection processes of a variety of different viruses and has revealed many of the common features and variant processes that are used (7-9, 33, 71).Here, we examine the uptake and infection of cells by parvovirus capsids and compare some of the steps followed by capsids that differ in their receptor binding properties and host ranges. Feline panleukopenia virus (FPV) infects cats (50, 66), binds the transferrin receptor-1 (TfR) on feline cells, and uses that receptor for uptake and infection (27, 44). FPV does not bind the canine TfR or infect dogs or cultured canine cells. Canine parvovirus (CPV) is a natural variant of FPV which emerged in 1978 after acquiring a small number of mutations that allow its capsid to bind the canine TfR (27). The original strain of CPV (designated CPV type 2 [CPV-2]) spread worldwide in dogs during 1978, but some of the same mutations that gave it the canine host range rendered it unable to infect cats (66, 67). CPV-2 was replaced worldwide during 1979 and 1980 by a natural variant, CPV type 2a (CPV-2a), which contained an additional four to five changes in its coat protein gene (48, 49). Subsequently, the canine viruses have continued to evolve, and additional single mutations have been selected that alter antigenic epitopes. Strains altered at VP2 residue 426 are designated CPV-2b (Asn426Asp) and CPV-2c (Asp426Glu) (13, 48). CPV-2a and its variants are able to infect both dogs and cats but show reduced binding to the feline TfR on cells and in vitro (27, 42). In addition, the affinity of binding to the canine TfR is much lower than that seen for the feline TfR (42).The TfR is a type II membrane protein expressed in nonlipid raft regions of the plasma membrane, and it binds iron-loaded (holo) transferrin (Tf) at neutral pH (2). TfR expression is tightly regulated, and it is more highly expressed on dividing cells with high iron needs, which would favor binding of these viruses. The TfRs of mice and humans are used as receptors for cell infection by the mouse mammary tumor virus and the New World hemorrhagic fever arenaviruses (52, 56).The TfR is assembled as a homodimer, and each monomer of the ectodomain is composed of protease-like, apical and helical domains, as well as a 30-Å membrane-proximal stalk (5, 20, 31). The transmembrane domain mediates membrane insertion and influences some aspects of trafficking within the cell, while the cytoplasmic domain contains a tyrosine-threonine-arginine-phenylalanine (YTRF) sequence that engages the clathrin-mediated endocytic machinery through AP-2 (adaptor protein-2) (53, 55). The TfR sequence also includes one or two cysteines adjacent to the inner leaflet of the membrane that may be palmitoylated to influence the rate of receptor recycling, and it also contains sequences that control basolateral localization in polarized cells (41). In the normal pathway of TfR-mediated entry, the TfR-holo-Tf complex is transported into the endosomal system, where low pH results in conformational changes and iron release. The TfR-Tf complex enters the early endosome, from which some of the complex is rapidly recycled to the cell surface while most passes to the perinuclear recycling endosome. From there it recycles to the cell surface where the iron-free apo-Tf is released at neutral pH (21, 22, 24, 37, 38, 69, 70). The rate of uptake and the efficiency of TfR recycling depend on the form of the ligand, and more than 97% of monomeric Tf recycles to the cell surface within 10 to 30 min. However, cross-linking TfRs with oligomeric Tf or antibodies causes the complexes to be retained within endosomes for longer times, and a higher proportion is trafficked to late endosomes and lysosomes for degradation (35).Holo-Tf binds the membrane-proximal side of the feline and canine TfR ectodomain (11), while virus binding involves the apical domain of the receptor as mutations in that structure affect the ability to bind FPV and CPV capsids (43) The feline and canine TfRs differ in ∼10% of their sequences, but a major difference controlling the CPV-specific binding is a unique glycosylation site in the apical domain of the canine TfR (43). Alteration of the glycosylated Asn to Lys (the feline TfR residue) allowed the canine TfR to bind FPV and also greatly increased the affinity of binding to CPV-2 and CPV-2a-related capsids (42).CPV and FPV have small (25 nm) nonenveloped capsids that package a single-stranded DNA genome of ∼5,120 bases (68). The particles are made up of two overlapping proteins, VP1 and VP2, with 90% of the capsid protein being VP2. VP1 contains a 143-residue amino (N)-terminal sequence that encodes a phospholipase A2 enzymatic activity, as well as basic amino acid motifs that play a role in nuclear localization (72). The VP1 unique region becomes exposed during cell entry without capsid disintegration, and the phospholipase A2 modifies the endosomal membrane to enhance endosomal escape (19, 75).Previous studies of cell entry by CPV, minute virus of mice, and various adeno-associated viruses (AAVs) show that viral uptake primarily occurs through clathrin-mediated endocytosis. However, when the AP-2-interacting sequences in the cytoplasmic tail of the feline TfR were mutated or deleted, the altered receptor still allowed CPV infection at a similar efficiency to that of wild-type TfR (26). The intracellular pathways of viral entry and trafficking have been examined by using cells fixed at various times after uptake and then antibody stained for virus and cellular markers or by expressing green fluorescent protein (GFP)-labeled markers. Time courses examined were between 1 and 6 h, and sequential steps of trafficking were suggested, with the virus passed from the early endosomes to the recycling endosome, followed by localization in late endosomes and lysosomes after uptake (65). By fluorescent antibody staining, VP1 release occurred only hours after uptake, possibly in a low-pH degradative compartment (64, 72). In addition, CPV capsids appear to remain associated with the receptor for 1 to 2 h after virus uptake as antibodies against the TfR cytoplasmic tail microinjected into feline CRFK cells block infection in this time period (44). Infection is also blocked by neutralizing the low pH of the endosomal system with ammonium chloride or bafilomycin A1, although it is not clear whether this is due to direct effects on the capsid or to indirect alterations in endosomal trafficking. When the X-ray crystal structures of capsids of CPV and FPV were determined at low pH or in the presence of EDTA or when capsids were examined for changes in protease susceptibility, only small changes in surface loops of the viral structure were present (40, 60).Here, we used microscopy to examine dynamic steps in the binding, uptake, and early trafficking of parvovirus capsids in live canine and feline cells. Labeled capsids were seen to undergo rapid movement into multiple endosomal compartments shortly after entry. Initial binding of CPV to canine cells involved filopodia while in feline cells the virus bound primarily to receptors on the cell body. In cells expressing GFP-conjugated Rab proteins, particles rapidly localized to multiple endosomal compartments in the cytoplasm after uptake, which gradually accumulated near the microtubule-organizing center. The distribution of intracellular viruses and the viral infectivity in feline cells were altered by expression of either constitutively active (CA) or dominant negative (DN) mutants of the Rab proteins.  相似文献   

12.
Polarized entry of canine parvovirus in an epithelial cell line.   总被引:3,自引:1,他引:2       下载免费PDF全文
The binding and uptake of canine parvovirus (CPV) in polarized epithelial cells were investigated by growing the cells on a permeable support and inoculating with the virus either from the apical or basolateral surface. Binding of radiolabeled CPV occurred preferentially on the basolateral surface. In contrast, when a similar experiment was carried out on nonpolarized A72 cells, virus binding occurred regardless of the direction of virus input. Binding appeared to be specific for CPV and could not be competitively inhibited by either bovine or porcine parvovirus. Analysis of the binding data revealed a high-affinity receptor (10(5) per cell) for CPV on the basolateral surfaces of MDCK cells (Kd, 29 pM). In indirect immunofluorescence studies, virus entered only from the basolateral surfaces of MDCK cells. These results provide evidence for a functional CPV-specific receptor that is expressed only on the basolateral surfaces of polarized epithelial cells, a result that has interesting consequences for viral pathogenesis.  相似文献   

13.
Determining how viruses infect new hosts via receptor-binding mechanisms is important for understanding virus emergence. We studied the binding kinetics of canine parvovirus (CPV) variants isolated from raccoons—a newly recognized CPV host—to different carnivore transferrin receptors (TfRs) using single-particle tracking. Our data suggest that CPV may utilize adhesion-strengthening mechanisms during TfR binding and that a single mutation in the viral capsid at VP2 position 300 can profoundly alter receptor binding and infectivity.  相似文献   

14.
Canine parvovirus disease is an acute infectious disease caused by canine parvovirus (CPV). Current commercial vaccines are mainly attenuated and inactivated; as such, problems concerning safety may occur. To resolve this problem, researchers developed virus-like particles (VLPs) as biological nanoparticles resembling natural virions and showing high bio-safety. This property allows the use of VLPs for vaccine development and mechanism studies of viral infections. Tissue-specific drug delivery also employs VLPs as biological nanomaterials. Therefore, VLPs derived from CPV have a great potential in medicine and diagnostics. In this study, small ubiquitin-like modifier (SUMO) fusion motif was utilized to express a whole, naturalVP2 protein of CPV in Escherichia coli. After the cleavage of the fusion motif, the CPV VP2 protein has self-assembled into VLPs. The VLPs had a size and shape that resembled the authentic virus capsid. However, the self-assembly efficiency of VLPs can be affected by different pH levels and ionic strengths. The mice vaccinated subcutaneously with CPV VLPs and CPV-specific immune responses were compared with those immunized with the natural virus. This result showed that VLPs can effectively induce anti-CPV specific antibody and lymphocyte proliferation as a whole virus. This result further suggested that the antigen epitope of CPV was correctly present on VLPs, thereby showing the potential application of a VLP-based CPV vaccine.  相似文献   

15.
Understanding the mechanisms of cross-species virus transmission is critical to anticipating emerging infectious diseases. Canine parvovirus type 2 (CPV-2) emerged as a variant of a feline parvovirus when it acquired mutations that allowed binding to the canine transferrin receptor type 1 (TfR). However, CPV-2 was soon replaced by a variant virus (CPV-2a) that differed in antigenicity and receptor binding. Here we show that the emergence of CPV involved an additional host range variant virus that has circulated undetected in raccoons for at least 24 years, with transfers to and from dogs. Raccoon virus capsids showed little binding to the canine TfR, showed little infection of canine cells, and had altered antigenic structures. Remarkably, in capsid protein (VP2) phylogenies, most raccoon viruses fell as evolutionary intermediates between the CPV-2 and CPV-2a strains, suggesting that passage through raccoons assisted in the evolution of CPV-2a. This highlights the potential role of alternative hosts in viral emergence.  相似文献   

16.
Canine parvovirus (CPV) is a small, nonenveloped virus that is a host range variant of a virus which infected cats and changes in the capsid protein control the ability of the virus to infect canine cells. We used a variety of approaches to define the early stages of cell entry by CPV. Electron microscopy showed that virus particles concentrated within clathrin-coated pits and vesicles early in the uptake process and that the infecting particles were rapidly removed from the cell surface. Overexpression of a dominant interfering mutant of dynamin in the cells altered the trafficking of capsid-containing vesicles. There was a 40% decrease in the number of CPV-infected cells in mutant dynamin-expressing cells, as well as a approximately 40% decrease in the number of cells in S phase of the cell cycle, which is required for virus replication. However, there was also up to 10-fold more binding of CPV to the surface of mutant dynamin-expressing cells than there was to uninduced cells, suggesting an increased receptor retention on the cell surface. In contrast, there was little difference in virus binding, virus infection rate, or cell cycle distribution between induced and uninduced cells expressing wild-type dynamin. CPV particles colocalized with transferrin in perinuclear endosomes but not with fluorescein isothiocyanate-dextran, a marker for fluid-phase endocytosis. Cells treated with nanomolar concentrations of bafilomycin A1 were largely resistant to infection when the drug was added either 30 min before or 90 min after inoculation, suggesting that there was a lag between virus entering the cell by clathrin-mediated endocytosis and escape of the virus from the endosome. High concentrations of CPV particles did not permeabilize canine A72 or mink lung cells to alpha-sarcin, but canine adenovirus type 1 particles permeabilized both cell lines. These data suggest that the CPV entry and infection pathway is complex and involves multiple vesicular components.  相似文献   

17.
The three-dimensional structure of expressed VP2 capsids of Aleutian mink disease parvovirus strain G (ADVG-VP2) has been determined to 22 A resolution by cryo-electron microscopy and image reconstruction techniques. A structure-based sequence alignment of the VP2 capsid protein of canine parvovirus (CPV) provided a means to construct an atomic model of the ADVG-VP2 capsid. The ADVG-VP2 reconstruction reveals a capsid structure with a mean external radius of 128 A and several surface features similar to those found in human parvovirus B19 (B19), CPV, feline panleukopenia virus (FPV), and minute virus of mice (MVM). Dimple-like depressions occur at the icosahedral twofold axes, canyon-like regions encircle the fivefold axes, and spike-like protrusions decorate the threefold axes. These spikes are not present in B19, and they are more prominent in ADV compared to the other parvoviruses owing to the presence of loop insertions which create mounds near the threefold axes. Cylindrical channels along the fivefold axes of CPV, FPV, and MVM, which are surrounded by five symmetry-related beta-ribbons, are closed in ADVG-VP2 and B19. Immunoreactive peptides made from segments of the ADVG-VP2 capsid protein map to residues in the mound structures. In vitro tissue tropism and in vivo pathogenic properties of ADV map to residues at the threefold axes and to the wall of the dimples.  相似文献   

18.
[目的]探索犬细胞毒性T细胞相关抗原-4(cytotoxic T lymphocyte-associated antigen-4,CTLA-4)胞外区作为免疫佐剂的可行性.[方法]根据已发表序列设计引物,用RT-PCR扩增CTLA-4胞外区编码序列,用PCR扩增犬细小病毒(canine parvovirus,CPV)VP2蛋白主要抗原表位基因片段VP2S,将VP2S克隆入含和不含CTLA-4胞外区基因片段的原核表达质粒pQE-31;用获得的重组质粒pQE-CTLA-4-VP2S和pQE-VP2S转化大肠杆菌,并进行诱导表达;用相同剂量的重组蛋白VP2S和CTLA-4-VP2S免疫小鼠.用间接ELISA和血凝抑制试验比较两个免疫组的抗体水平.[结果]经过30次循环PCR扩增后,琼脂糖凝胶电泳显示预期大小的扩增产物;序列测定结果显示,克隆的毕格犬CTLA-4胞外区与已发表序列的核苷酸同源性为99.2%,氨基酸序列同源性为98.4%,结合B7分子的六肽基序(MYPPPY)无变化:VP2S与已发表CPV VP2的核苷酸序列同源性为99%,氨基酸序列同源性为98.6%:经IPTG诱导后,两种重组大肠杆菌表达预期的29kDa VP2S和42kDaCTLA-4-VP2S重组蛋白,两者均能被CPV抗血清识别;间接ELISA和血凝抑制试验结果显示,CTLA-4-VP2S免疫组的抗体产生时间为初免后第2周,抗体高峰期为初免后第4周,而VP2S免疫组的抗体产生时间为初免后第4周,抗体高峰期为初免后第5周,两个试验组高峰期ELISA抗体效价和血凝抑制抗体效价分别相差100倍和10倍.[结论]犬CTLA-4胞外区可作为分子佐剂促进CPV VP2蛋白抗体的产生.  相似文献   

19.
The adaptation of viruses to new hosts is a poorly understood process likely involving a variety of viral structures and functions that allow efficient replication and spread. Canine parvovirus (CPV) emerged in the late 1970s as a host-range variant of a virus related to feline panleukopenia virus (FPV). Within a few years of its emergence in dogs, there was a worldwide replacement of the initial virus strain (CPV type 2) by a variant (CPV type 2a) characterized by four amino acid differences in the capsid protein. However, the evolutionary processes that underlie the acquisition of these four mutations, as well as their effects on viral fitness, both singly and in combination, are still uncertain. Using a comprehensive experimental analysis of multiple intermediate mutational combinations, we show that these four capsid mutations act in concert to alter antigenicity, cell receptor binding, and relative in vitro growth in feline cells. Hence, host adaptation involved complex interactions among both surface-exposed and buried capsid mutations that together altered cell infection and immune escape properties of the viruses. Notably, most intermediate viral genotypes containing different combinations of the four key amino acids possessed markedly lower fitness than the wild-type viruses.  相似文献   

20.
A mammalian baculovirus delivery system was developed to study targeting in Norden Laboratories feline kidney (NLFK) cells of the capsid proteins of canine parvovirus (CPV), VP1 and VP2, or corresponding counterparts fused to EGFP. VP1 and VP2, when expressed alone, both had equal nuclear and cytoplasmic distribution. However, assembled form of VP2 had a predominantly cytoplasmic localization. When VP1 and VP2 were simultaneously present in cells, their nuclear localization increased. Thus, confocal immunofluorescence analysis of cells transduced with the different baculovirus constructs or combinations thereof in the absence or presence of infecting CPV revealed that the VP1 protein is a prerequisite for efficient targeting of VP2 to the nucleus. The baculovirus vectors were functional and the genes of interest efficiently introduced to this CPV susceptible mammalian cell line. Thus, we show evidence that the system could be utilized to study targeting of the CPV capsid proteins.  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