首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
2.
3.
The pSub201-pAAV/Ad plasmid cotransfection system was developed to eliminate homologous recombination which leads to generation of the wild-type (wt) adeno-associated virus type 2 (AAV) during recombinant vector production. The extent of contamination with wt AAV has been documented to range between 0.01 and 10%. However, the precise mechanism of generation of the contaminating wt AAV remains unclear. To characterize the wt AAV genomes, recombinant viral stocks were used to infect human 293 cells in the presence of adenovirus. Southern blot analyses of viral replicative DNA intermediates revealed that the contaminating AAV genomes were not authentic wt but rather wt AAV-like sequences derived from recombination between (i) AAV inverted terminal repeats (ITRs) in the recombinant plasmid and (ii) AAV sequences in the helper plasmid. Replicative AAV DNA fragments, isolated following amplification through four successive rounds of amplification in adenovirus-infected 293 cells, were molecularly cloned and subjected to nucleotide sequencing to identify the recombinant junctions. Following sequence analyses of 31 different ends of AAV-like genomes derived from two different recombinant vector stocks, we observed that all recombination events involved 10 nucleotides in the AAV D sequence distal to viral hairpin structures. We have recently documented that the first 10 nucleotides in the D sequence proximal to the AAV hairpin structures are essential for successful replication and encapsidation of the viral genome (X.-S. Wang et al., J. Virol. 71:3077–3082, 1997), and it was noteworthy that in each recombinant junction sequenced, the same 10 nucleotides were retained. We also observed that adenovirus ITRs in the helper plasmid were involved in illegitimate recombination with AAV ITRs, deletions of which significantly reduced the extent of wt AAV-like particles. Furthermore, the combined use of recombinant AAV plasmids lacking the distal 10 nucleotides in the D sequence and helper plasmids lacking the adenovirus ITRs led to complete elimination of replication-competent wt AAV-like particles in recombinant vector stocks. These strategies should be useful in producing clinical-grade AAV vectors suitable for human gene therapy.  相似文献   

4.
5.
The single-stranded adeno-associated virus (AAV) genome is flanked by terminal hairpinned origins of DNA replication (terminal repeats [TRs]) that are nicked at the terminal resolution site (trs) by the AAV Rep protein in an ATP-dependent, site-specific manner. Here we determine the minimal trs sequence necessary for Rep cleavage, 3'-CCGGT/TG-5', and show that this 7-base core sequence is required only on the nicked strand. We also identify a potential stem-loop structure at the trs. Interestingly, Rep nicking on a TR substrate that fixes this trs stem-loop in the extruded form no longer requires ATP. This suggests that ATP-dependent Rep helicase activity is necessary to unwind the duplex trs and extrude the stem-loop structure, prior to the ATP-independent Rep transesterification reaction. The extrusion of origin stem-loop structures prior to nicking appears to be a general mechanism shared by plant and animal viruses and bacterial plasmids. In the case of AAV, this mechanism of TR nicking would provide a possible regulatory function.  相似文献   

6.
The nonstructural adeno-associated virus type 2 Rep proteins are known to control viral replication and thus provide the single-stranded DNA genomes required for packaging into preformed capsids. In addition, complexes between Rep proteins and capsids have previously been observed in the course of productive infections. Such complexes have been interpreted as genome-linked Rep molecules associated with the capsid upon successful DNA encapsidation. Here we demonstrate via coimmunoprecipitation, cosedimentation, and yeast two-hybrid analyses that the Rep-VP association also occurs in the absence of packageable genomes, suggesting that such complexes could be involved in the preparation of empty capsids for subsequent encapsidation steps. The Rep domain responsible for the observed Rep-VP interactions is situated within amino acids 322 to 482. In the presence of all Rep proteins, Rep52 and, to a lesser extent, Rep78 are most abundantly recovered with capsids, whereas Rep68 and Rep40 vary in association depending on their expression levels. Rep78 and Rep52 are bound to capsids to roughly the same extent as the minor capsid protein VP2. Complexes of Rep78 and Rep52 with capsids differ in their respective detergent stabilities, indicating that they result from different types of interactions. Rep-VP interaction studies suggest that Rep proteins become stably associated with the capsid during the assembly process. Rep-capsid complexes can reach even higher complexity through additional Rep-Rep interactions, which are particularly detergent labile. Coimmunoprecipitation and yeast two-hybrid data demonstrate the interaction of Rep78 with Rep68, of Rep68 with Rep52, and weak interactions of Rep40 with Rep52 and Rep78. We propose that the large complexes arising from these interactions represent intermediates in the DNA packaging pathway.  相似文献   

7.
The human parvovirus adeno-associated virus (AAV) infects a broad range of cell types, including human, nonhuman primate, canine, murine, and avian. Although little is known about the initial events of virus infection, AAV is currently being developed as a vector for human gene therapy. Using defined mutant CHO cell lines and standard biochemical assays, we demonstrate that heparan sulfate proteoglycans mediate both AAV attachment to and infection of target cells. Competition experiments using heparin, a soluble receptor analog, demonstrated dose-dependent inhibition of AAV attachment and infection. Enzymatic removal of heparan but not chondroitin sulfate moieties from the cell surface greatly reduced AAV attachment and infectivity. Finally, mutant cell lines that do not produce heparan sulfate proteoglycans were significantly impaired for both AAV binding and infection. This is the first report that proteoglycan has a role in cellular attachment of a parvovirus. Together, these results demonstrate that membrane-associated heparan sulfate proteoglycan serves as the viral receptor for AAV type 2, and provide an explanation for the broad host range of AAV. Identification of heparan sulfate proteoglycan as a viral receptor should facilitate development of new reagents for virus purification and provide critical information on the use of AAV as a gene therapy vector.  相似文献   

8.
The adeno-associated virus (AAV) is unique in its ability to target viral DNA integration to a defined region of human chromosome 19 (AAVS1). Since AAVS1 sequences are not conserved in a rodent’s genome, no animal model is currently available to study AAV-mediated site-specific integration. We describe here the generation of transgenic rats and mice that carry the AAVS1 3.5-kb DNA fragment. To test the response of the transgenic animals to Rep-mediated targeting, primary cultures of mouse fibroblasts, rat hepatocytes, and fibroblasts were infected with wild-type wt AAV. PCR amplification of the inverted terminal repeat (ITR)-AAVS1 junction revealed that the AAV genome integrated into the AAVS1 site in fibroblasts and hepatocytes. Integration in rat fibroblasts was also observed upon transfection of a plasmid containing the rep gene under the control of the p5 and p19 promoters and a dicistronic cassette carrying the green fluorescent protein (GFP) and neomycin (neo) resistance gene between the ITRs of AAV. The localization of the GFP-Neo sequence in the AAVS1 region was determined by Southern blot and FISH analysis. Lastly, AAV genomic DNA integration into the AAVS1 site in vivo was assessed by virus injection into the quadriceps muscle of transgenic rats and mice. Rep-mediated targeting to the AAVS1 site was detected in several injected animals. These results indicate that the transgenic lines are proficient for Rep-mediated targeting. These animals should allow further characterization of the molecular aspects of site-specific integration and testing of the efficacy of targeted integration of AAV recombinant vectors designed for human gene therapy.  相似文献   

9.
Adeno-associated virus (AAV) is a human parvovirus that replicates only in cells coinfected with a helper virus, such as adenovirus or herpes simplex virus type 1 (HSV-1). We previously showed that nine HSV-1 factors are able to support AAV rep gene expression and genome replication. To elucidate the strategy of AAV replication in the presence of HSV-1, we undertook a proteomic analysis of cellular and HSV-1 factors associated with Rep proteins and thus potentially recruited within AAV replication compartments (AAV RCs). This study resulted in the identification of approximately 60 cellular proteins, among which factors involved in DNA and RNA metabolism represented the largest functional categories. Validation analyses indicated that the cellular DNA replication enzymes RPA, RFC, and PCNA were recruited within HSV-1-induced AAV RCs. Polymerase δ was not identified but subsequently was shown to colocalize with Rep within AAV RCs even in the presence of the HSV-1 polymerase complex. In addition, we found that AAV replication is associated with the recruitment of components of the Mre11/Rad50/Nbs1 complex, Ku70 and -86, and the mismatch repair proteins MSH2, -3, and -6. Finally, several HSV-1 factors were also found to be associated with Rep, including UL12. We demonstrated for the first time that this protein plays a role during AAV replication by enhancing the resolution of AAV replicative forms and AAV particle production. Altogether, these analyses provide the basis to understand how AAV adapts its replication strategy to the nuclear environment induced by the helper virus.Adeno-associated virus (AAV) is a human parvovirus that is currently used as a gene transfer vector (14). AAV particles consist of a small icosahedral capsid protecting a single 4.7-kb single-stranded DNA (ssDNA) genome with two open reading frames, rep and cap, surrounded by inverted terminal repeats (ITRs). The ITRs are the only sequences required in cis for genome replication and packaging. The rep gene encodes four nonstructural Rep proteins: Rep78, -68, -52, and -40. The two larger isoforms, Rep78 and -68, have origin binding, helicase, and site-specific endonuclease activities and are involved in AAV gene expression and genome processing, including replication and site-specific integration (39). The two smaller Rep isoforms are not required for AAV DNA replication but are involved in the control of viral gene expression and packaging of viral DNA (30).When wild-type (wt) AAV infects a cell in the absence of a helper virus, it enters latency. Latent AAV genomes persist in cells either as episomes or as integrated genomes, preferentially at a specific locus (named AAVS1) on human chromosome 19. In most instances, no detectable viral gene expression or genome replication occurs unless the cell is co- or superinfected by a helper virus, such as adenovirus, herpes simplex virus type 1 (HSV-1), or HSV-2. Under these conditions, AAV replication and assembly take place in large intranuclear domains called replication compartments (RCs) that frequently colocalize with replication domains formed by the helper virus itself (81). The viral genome replicates by leading-strand synthesis and generates new ssDNA molecules by a strand displacement mechanism that occurs after strand- and site-specific cleavage of viral DNA by Rep78/68 within the ITRs (39).Studies conducted on the relationship between AAV and its helper viruses are important not only to identify helper activities that can be used to produce recombinant AAV vectors but also to understand how AAV adapts its replication strategy to the helper virus and to the nuclear environment in general. Adenovirus helper functions have historically been the first and most extensively studied functions. These studies have shown that adenovirus helps AAV by stimulating viral gene expression and by enhancing AAV genome replication, mostly indirectly (19). Indeed, early studies showed that the adenovirus polymerase (E2b) is dispensable for AAV replication (8) and that the viral DNA-binding protein (DBP), the product of the E2a gene, is able to modestly enhance the processivity of AAV genome replication in vitro (77). More recently, the adenovirus proteins E1b55k and E4orf6 were shown to stimulate AAV genome replication by degrading the cellular Mre11/Rad50/Nbs1 (MRN) complex that restricts AAV genome replication during adenovirus coinfection (32). The concept that AAV genome replication can rely mostly, if not uniquely, on direct help from cellular factors was further strengthened by the demonstration that purified proteins such as replication protein A (RPA), replication factor C (RFC), proliferating cell nuclear antigen (PCNA), minichromosome maintenance (MCM) proteins, and DNA polymerase δ (Pol δ) were sufficient to replicate the AAV genome in vitro in the presence of Rep (40-41, 43). The involvement of these cellular proteins during AAV genome replication was also confirmed by the proteomic analysis of factors associated with Rep proteins during adenovirus-induced AAV replication (42).Interestingly, studies conducted on HSV-1 helper activities suggest that the strategy of AAV replication may vary depending on the helper virus. Indeed, previous studies showed that the HSV-1 helicase-primase (HP) complex (UL5/8/52) and DBP (ICP8) could replicate transfected AAV-2 plasmids (80) and that the helicase activity, but not primase activity, of the HP complex was required for this effect (62, 66). More recently, a comprehensive study of HSV-1 helper activities demonstrated that the HSV-1 immediate-early proteins ICP0, ICP4, and ICP22 could stimulate rep gene expression, probably by diminishing intrinsic antiviral effects (1, 18). In addition, the HSV-1 DNA polymerase encoded by UL30, along with its associated processivity factor (UL42), although not strictly required, was demonstrated to significantly increase AAV replication levels induced in the presence of the HP complex and ICP8. Interestingly, the HSV-1 HP complex, DBP, and polymerase were also shown to be sufficient to replicate AAV DNA in vitro in the presence of Rep proteins without any cellular protein (78). Altogether, these observations indicate that in the context of an HSV-1 coinfection, AAV relies extensively on viral activities provided by the helper that directly participate in AAV genome replication.To further elucidate the strategy of AAV replication in the presence of HSV-1, we undertook a proteomic analysis to identify the cellular and HSV-1 factors associated with Rep proteins and, consequently, potentially recruited within AAV RCs. To analyze Rep-associated proteins in the presence and absence of HSV-1 DNA replication, this analysis was performed using wt HSV-1 and an HSV-1 mutant in which the DNA polymerase encoded by the UL30 gene is absent (HSVΔUL30). This study resulted in the identification of approximately 60 cellular proteins, among which the largest functional categories corresponded to factors involved in DNA and RNA metabolism. Immunofluorescence analyses confirmed that in the presence of HSV-1, a basal set of cellular DNA replication enzymes, including RPA, RFC, and PCNA, was recruited within AAV RCs, with the exception of the MCM helicases. The cellular DNA polymerases, in particular Pol δ, were not identified by this analysis but subsequently were shown to be recruited in AAV RCs even in the presence of the HSV-1 polymerase complex. In addition, our results indicate that AAV replication induced by HSV-1 is associated with the recruitment of DNA repair factors, including components of the MRN complex, the Ku proteins, PARP-1, and factors of the mismatch repair (MMR) pathway. Finally, several HSV-1 proteins, most notably the UL12 protein, were also identified within AAV RCs. Our analyses confirmed the association between UL12 and Rep and demonstrated for the first time that this viral exonuclease plays a critical role during AAV replication by enhancing the formation of discrete AAV replicative forms and the production of AAV particles.Altogether, these results indicate that in the presence of HSV-1, AAV may replicate by using a basal set of cellular DNA replication enzymes but also relies extensively on HSV-1-derived proteins for its replication, including UL12, a newly discovered helper factor. These results suggest that AAV may be able to differentially adapt its replication strategy to the nuclear environment induced by the helper virus.  相似文献   

10.
Cloning and Characterization of Adeno-Associated Virus Type 5   总被引:20,自引:8,他引:12       下载免费PDF全文
Adeno-associated virus type 5 (AAV5) is distinct from other dependovirus serotypes based on DNA hybridization and serological data. To better understand the biology of AAV5, we have cloned and sequenced its genome and generated recombinant AAV5 particles. The single-stranded DNA genome is similar in length and genetic organization to that of AAV2. The rep gene of AAV5 is 67% homologous to AAV2, with the majority of the changes occurring in the carboxyl and amino termini. This homology is much less than that observed with other reported AAV serotypes. The inverted terminal repeats (ITRs) are also unique compared to those of the other AAV serotypes. While the characteristic AAV hairpin structure and the Rep DNA binding site are retained, the consensus terminal resolution site is absent. These differences in the Rep proteins and the ITRs result in a lack of cross-complementation between AAV2 and AAV5 as measured by the production of recombinant AAV particles. Alignment of the cap open reading frame with that of the other AAV serotypes identifies both conserved and variable regions which could affect tissue tropism and particle stability. Comparison of transduction efficiencies in a variety of cells lines and a lack of inhibition by soluble heparin indicate that AAV5 may utilize a distinct mechanism of uptake compared to AAV2.  相似文献   

11.
The site-specific integration of wild-type adeno-associated virus (wtAAV) into the human genome is a very attractive feature for the development of AAV-based gene therapy vectors. However, knowledge about integration of wtAAV, as well as currently configured recombinant AAV (rAAV) vectors, is limited. By using a modified Alu-PCR technique to amplify and sequence the vector-cellular junctions, we provide the first direct evidence both in vitro and in vivo of rAAV-mediated transgene integration in several types of nondividing cells, including neurons. This novel technique will be highly useful for further delineating the mechanisms underlying AAV-mediated integration, including issues of frequency, site preference, and DNA rearrangement in human as well as animal cells. Results from these studies should be beneficial for the development of the next generation of gene delivery vectors.  相似文献   

12.
A recent clinical trial in patients with hemophilia B has suggested that adeno-associated virus (AAV) capsid-specific cytotoxic T lymphocytes (CTLs) eliminated AAV-transduced hepatocytes and resulted in therapeutic failure. AAV capsids elicit a CTL response in animal models; however, these capsid-specific CTLs fail to kill AAV-transduced target cells in mice. To better model the human clinical trial data in mice, we introduced an immunodominant epitope derived from ovalbumin (OVA; SIINFEKL) into the AAV capsid and tested CTL-mediated killing of AAV2-transduced target tissues in vivo. Initially, in vitro experiments demonstrated both classical class I and cross-presentation of the OVA antigen, following endogenous expression or AAV2-OVA vector transduction, respectively. Furthermore, an OVA-specific CTL response was elicited after muscular or systemic injection of the AAV2-OVA vector. Finally, CTL reactivity was enhanced in mice with established SIINFEKL-specific immunity after AAV2-OVA/α1 anti-trypsin (AAT) administration. Most importantly, these OVA-specific CTLs decreased AAT expression in mice treated with AAV2-OVA/AAT vector that followed a time course mimicking uncoating kinetics of AAV2 transduction in OVA-immunized mice. These results demonstrate that AAV capsid-derived antigens elicit CD8+ CTL reactivity, and these CTLs eliminated AAV-transduced target cells in mice. Notably, this model system can be exploited to study the kinetics of capsid presentation from different serotypes of AAV and permit the design of novel strategies to block CTL-mediated killing of AAV-transduced cells.Adeno-associated virus (AAV) is a single-stranded DNA parvovirus. Its replication relies on coinfection of a helper virus such as adenovirus or herpesvirus. In the absence of a helper virus, AAV establishes latency to integrate into the AAVS1 site of host chromosome 19 (11). The genome of AAV is ∼4.7 kb and contains two open reading frames encoding replication proteins and structural capsid proteins (21). The capsid proteins (VP) are composed of VP1, VP2, and VP3. The VP3 protein is the major structural component and constitutes nearly 80% of the virion shell with an overall ratio of 1:1:8 for VP1, VP2, VP3, respectively. While VP2 is thought to be nonessential for AAV transduction (30), the VP1 subunit contains a phospholipase A2 domain required for infectivity (9). Recombinant AAV (rAAV) vectors require only the 145-bp terminal repeats of the AAV genome in cis and all other viral factors supplied in trans for production (18). rAAV vectors have rapidly gained popularity in gene therapy applications and have proven effective in preclinical studies/clinical trials for a number of diseases (20, 31, 33).AAV vectors mount a potent humoral immune response against capsid in animals and human. However, AAV vectors only contain the therapeutic gene flanked by two 145-bp AAV terminal repeats devoid of any AAV genes(23). In addition, AAV initiates long-term stable therapeutic gene expression in animal models (3-5, 17, 31). Based on these observations AAV has been thought to be relatively nonimmunogenic regarding the induction of cytotoxic T lymphocytes (CTLs) specific for capsid proteins. In spite of all of these observations, the recent clinical trial for hemophilia B (F9) gene therapy has otherwise suggested that AAV2 capsid initiates cell-mediated immunity that eliminates the AAV2 encoding F9 (AAV2/F9) vector transduced liver cells (15). Against this backdrop, numerous attempts to replicate aforementioned observations in animal models have been made. Preliminary results from these studies support direct presentation and cross-presentation of the AAV2 capsid in animal models (6, 12, 13, 22, 29). However, capsid-specific CTLs did not eliminate AAV2-transduced target cells in mice (12, 13, 29), inconsistent with observations made in a clinical trial for hemophilia B with AAV2/F9 gene therapy. A potential explanation for this discrepancy is the weak immunogenicity of the AAV2 capsid in mice. Accordingly, we hypothesized that incorporation of a peptide epitope into the AAV2 capsid would increase immunogenicity of the rAAV and therefore could be exploited to mimic events ongoing in humans and study approaches to block capsid-specific CTL reactivity in mice.We chose to introduce the MHC-H2Kb-restricted SIINFEKL peptide derived from ovalbumin (OVA) into AAV2 capsid. Integration of the OVA epitope into AAV capsids elicited a specific CTL response. Most importantly, after administration of genetically engineered AAV2 vectors into OVA peptide-immunized mice, OVA-specific CTL reactivity was further enhanced, thereby limiting transgene expression in vivo. The modified vector described herein is a potentially valuable tool for future studies focused on developing strategies to evade capsid-specific CTL-mediated elimination of AAV-transduced target cells in animal models.  相似文献   

13.
Human T-lymphotropic virus type 1 (HTLV-1) envelope (Env) glycoprotein mediates binding of the virus to its receptor on the surface of target cells and subsequent fusion of virus and cell membranes. To better understand the mechanisms that control HTLV-1 Env trafficking and activity, we have examined two protein-protein interaction motifs in the cytoplasmic domain of Env. One is the sequence YSLI, which matches the consensus YXXΦ motifs that are known to interact with various adaptor protein complexes; the other is the sequence ESSL at the C terminus of Env, which matches the consensus PDZ-binding motif. We show here that mutations that destroy the YXXΦ motif increased Env expression on the cell surface and increased cell-cell fusion activity. In contrast, mutation of the PDZ-binding motif greatly diminished Env expression in cells, which could be restored to wild-type levels either by mutating the YXXΦ motif or by silencing AP2 and AP3, suggesting that interactions with PDZ proteins oppose an Env degradation pathway mediated by AP2 and AP3. Silencing of the PDZ protein hDlg1 did not affect Env expression, suggesting that hDlg1 is not a binding partner for Env. Substitution of the YSLI sequence in HTLV-1 Env with YXXΦ elements from other cell or virus membrane-spanning proteins resulted in alterations in Env accumulation in cells, incorporation into virions, and virion infectivity. Env variants containing YXXΦ motifs that are predicted to have high-affinity interaction with AP2 accumulated to lower steady-state levels. Interestingly, mutations that destroy the YXXΦ motif resulted in viruses that were not infectious by cell-free or cell-associated routes of infection. Unlike YXXΦ, the function of the PDZ-binding motif manifests itself only in the producer cells; AP2 silencing restored the incorporation of PDZ-deficient Env into virus-like particles (VLPs) and the infectivity of these VLPs to wild-type levels.Human T-lymphotropic virus type 1 (HTLV-1) envelope (Env), like most retroviral envelopes, is synthesized as a precursor protein in the endoplasmic reticulum, forms trimers, and is cleaved by a cellular furin-like protease as it transits through the trans-Golgi network on its way to the plasma membrane (7, 21, 31). Cleavage of the HTLV-1 Env precursor generates a 46-kDa surface subunit (SU, gp46) and a 21-kDa transmembrane protein (TM, gp21) (8, 43). SU contains the receptor-binding domain and is linked by a disulfide bond to TM, which anchors Env to the membrane and mediates fusion of virus and cell membranes after receptor engagement (11, 28, 40, 51). TM consists of extracellular, membrane-spanning, and cytoplasmic domains (31); the last contains motifs that direct Env trafficking, membrane targeting, and virion incorporation. HTLV-1 is poorly transmitted as cell-free virus, and there is good evidence supporting a model in which virions are transmitted in a polarized fashion between lymphocytes that are in close contact (22, 30). Unlike murine leukemia virus (MLV) and Mason-Pfizer monkey virus (MPMV) Envs, in which the cytoplasmic domain (CD) is cleaved by the virus-encoded protease to activate fusogenic activity (3, 6, 19, 42), the HTLV-1 Env cytoplasmic domain is not cleaved and HTLV-1 Env exists on the cell surface in a highly fusogenic state. In many respects, HTLV-1 Env resembles versions of MLV or MPMV Envs that lack C-terminal amino acids, e.g., with elevated cell-cell fusion activity and low virion infectivity. It is not exactly clear how HTLV-1 Env is controlled such that virus infection can proceed without cell-cell fusion, but it is probable that Env trafficking plays an important role. The cytoplasmic domain of HTLV-1 Env is relatively short and contains two important trafficking motifs: a YXXΦ motif (YSLI), which is involved in membrane protein trafficking and basolateral sorting in polarized epithelial cells (10), and a PDZ-binding motif (ESSL), which can interact with numerous PDZ proteins but is not found in other retroviral Envs (2).The tyrosine-based sorting motif (YXXΦ, where Y is tyrosine, X is any amino acid, and Φ is a bulky hydrophobic amino acid) determines the trafficking and turnover of many membrane-spanning proteins in the cell (5, 39) and is present in most retroviral Env proteins (7). The YXXΦ motif interacts with the μ subunit of the heterotetrameric adaptor protein complexes AP1, AP2, AP3, and AP4. Each adaptor complex is involved in a specific trafficking pathway: AP1 and AP4 deliver cargo from the trans-Golgi network to the plasma membrane (13, 33, 48), AP2 directs the endocytosis of proteins from the cell surface, and AP3 is involved in lysosomal sorting (5, 12, 24, 35). Each type of μ subunit interacts with a distinct but overlapping type of tyrosine-based motif; the tyrosine and the Φ residues are most critical, but affinity is determined in large part by the variable amino acids at positions +1 and +2 relative to tyrosine and also by surrounding amino acids (5, 37). Furthermore, interactions between AP2 and the YXXΦ motif may be regulated by phosphorylation of μ2 (38, 47), by localized changes in phosphoinositide concentration, or by interactions between AP2 and docking factors (47). Although most retroviral Env proteins contain YXXΦ-sorting motifs, the sequences of the motifs and their roles in Env trafficking and function appear to vary widely among different retroviruses. For example, mutation of the YXXΦ motif in MLV Env interferes with basolateral targeting of Env and diminishes viral pathogenesis in vivo but has little effect on Env accumulation at the plasma membrane (9, 16, 23, 25, 29). Mutations in the YXXΦ motif in MPMV Env are similar to those in MLV Evn and also were reported to affect Env incorporation into virions (45). Mutation of the YXXΦ motif in HTLV-1 Env was previously shown to decrease Env endocytosis, increase cell-cell fusion, increase Env incorporation into virions, abolish basolateral targeting, and decrease virus infectivity (1, 10).The most abundant protein-protein interaction domains in mammalian cells are the PDZ domains; more than 400 PDZ proteins are encoded in the human genome. PDZ domains are modular, recognize short C-terminal peptide motifs, and are often found in multiple copies or in combination with other protein interaction domains (36, 46, 50). PDZ proteins have the ability to form supramolecular scaffolds that coordinate signaling, synapse formation, cell polarity, and trafficking of interacting proteins (26, 44, 53). With respect to the last, it is important to note that PDZ proteins can delay the internalization of G protein-coupled receptors, ion channels, and membrane transporters (17, 41, 49, 52). Among retroviral Env proteins, only HTLV and simian T-lymphotropic virus (STLV) Envs contain putative PDZ-binding motifs. A yeast two-hybrid screen using the HTLV-1 Env cytoplasmic domain (CD) as bait identified the PDZ protein hDlg (human homolog of disc large protein) as a potential binding partner (2). In vitro pulldown experiments showed that a glutathione S-transferase (GST)-EnvCD fusion protein interacted with several PDZ proteins from cell lysates, one of which was hDlg. In one study, mutation of the PDZ-binding motif in HTLV-1 Env inhibited cell-cell fusion (2); in another study, hDlg small interfering RNA (siRNA) silencing caused a modest reduction in syncytium formation (54). Neither study examined how the PDZ-binding motif controls Env expression, membrane targeting, trafficking, or virus infectivity. Thus, it is still unclear which PDZ proteins interact with HTLV-1 Env in vivo and how those interactions affect Env trafficking and activity.In this paper, functional interactions between the YXXΦ motif and the PDZ-binding motif in the cytoplasmic domain of HTLV-1 Env were investigated by mutagenesis of Env and by siRNA silencing of potential cellular interacting proteins. The YXXΦ motif in HTLV-1 Env appears to interact primarily with AP2 and AP3, which regulate Env endocytosis and lysosomal degradation, respectively. Mutations that ablated the YXXΦ motif increased Env accumulation on the cell surface. The PDZ-binding motif at the C terminus of Env appears to delay Env turnover. Mutation of the PDZ-binding element diminished Env accumulation in cells to very low levels, indicating that loss of the PDZ-binding motif accelerates Env degradation. Expression of Env with a mutated PDZ-binding motif could be restored to normal levels by also mutating the YXXΦ motif or by silencing AP2 or AP3. The ability of the PDZ-binding motif to alter the activity of the YXXΦ motif depends on the particular sequence of the latter. The attenuating effect of the PDZ-binding motif on Env endocytosis could be overcome by substitution of the YSLI motif in HTLV-1 Env with YXXΦ elements from other cell or virus proteins that are predicted to have higher affinities for AP2 than the YSLI motif of HTLV-1 Env.  相似文献   

14.
Five site-specific adeno-associated virus integrants generated in a model system with an Epstein-Barr virus-based shuttle vector have been characterized. The results suggest a deletion-substitution mechanism of recombination.  相似文献   

15.
Adeno-associated virus type 2 (AAV 2) is the only eukaryotic virus capable of site-specific integration; the target site is at chromosome 19q13.4, a site termed AAVS1. The biology of AAV latency has been extensively studied in cell culture, yet the precise mechanism and the required cellular factors are not known. In this study, we assessed the relative frequencies of stable site-specific integration by characterization of cell clones containing integrated AAV vectors. By this assay, two proteins involved in nonhomologous end joining (NHEJ), DNAPKcs and ligase IV, exhibit differential effects on AAV site-specific integration. DNAPKcs is not required; its presence increases the frequency of junction formation indicative of site-specific integration, but seems to reduce the ratio of site-specific integration to random integration (i.e., the latter is even more enhanced). In contrast, site-specific integration is significantly reduced relative to random integration in cells deficient in ligase IV expression. Furthermore, we show that single-stranded AAV vectors are better substrates for site-specific integration than are self-complementary AAV vectors; the absence of DNAPKcs did not affect the targeted integration of these double-stranded AAV vectors. Together, these data suggest that NHEJ proteins participate in site-specific integration, and indicate a role for the single-stranded form of AAV DNA in targeted integration.Adeno-associated virus (AAV) is a ubiquitous human virus (∼80% of adults are seropositive [4]); like other nuclear DNA viruses, it causes persistent infections. Productive infection by AAV requires coinfection with either an adenovirus or herpesvirus (1, 21). In cell culture, an AAV latent infection is readily established by infection with a high multiplicity of infection (MOI) (>250 infectious particles/cell) in the absence of a helper virus coinfection, and such latent infections have been reported to persist for over 100 passages (3). Because of the stability of latent infections, it has been possible to clone latently infected cells and to determine the molecular characteristics of the persistent viral genome. More than 65 to 90% of such clones have been determined to have the AAV genome integrated at a specific site on chromosome 19q13.4 (16, 26). The degree of specificity of the integration site is unique among human viruses. Integration is not specific at the nucleotide level, but a specific target sequence has been determined, which includes a binding site for the AAV Rep protein (RBS) and a so-called terminal resolution site (TRS) which is cleaved in one strand by the Rep protein during AAV DNA replication (19). In addition to the target site, which has been termed AAVS1, site-specific integration has been demonstrated to require the AAV rep protein (either Rep 68 or 78) and a sequence in the inverted terminal repeat homologous to AAVS1 (31, 33). A third Rep binding site (RBS) is found in the promoter at map position 5, which has been reported to greatly enhance site-specific integration (24).Although there have been numerous studies of the mechanisms involved in site-specific integration, many aspects remain to be elucidated. In particular, when during the cell cycle site-specific integration occurs is unknown, and the cellular proteins which are involved have not been identified. Such integration might occur as the consequence of nonhomologous end joining (NHEJ) or homologous recombination (HR). Although the former seems to be a more likely pathway, because integration is into a region of very limited homology, the fact that there is some homology between the minimal essential target sequence and the AAV inverted terminal repeat (ITR) suggests that homologous recombination cannot be arbitrarily dismissed as a possibility. Another unknown feature of site-specific integration is the molecular state of the AAV substrate, i.e., whether the substrate is single or double stranded. Transfection of AAV-containing plasmids does lead to site-specific integration (24); thus, a circular form of duplex AAV DNA can serve as the initial substrate, although it has been more challenging to detect significant levels of such integration after transfection with linear duplex AAV DNA (J. Dyall and K. I. Berns, unpublished data).There have been several studies of the fate of AAV vectors used in gene therapy. The studies have been performed primarily in mice and on occasion in vitro. AAV vectors lack the Rep gene and thus do not preferentially integrate into AAVS1 (25). However, random integration at a low frequency, less than 10−7 (5), does take place and is of concern because of the potential for insertional mutagenesis and the attendant possibility of oncogenesis (20, 22). Such studies have indicated that NHEJ is involved and that different tissues seem to have variable capacities to support integration (14). Other studies have studied recombination between the ITRs at the ends of AAV DNA. Of particular note have been such studies of AAV vectors containing self-complementary genomes (8). These genomes contain complementary sequences separated by an ITR sequence, which itself is a hairpin sequence. Consequently, these genomes can hairpin on themselves to form a duplex structure which contains three hairpinned ITRs, the one in the middle and the two on the ends. The hairpinned ends are substrates for recombination mediated by NHEJ.If NHEJ were involved in AAV integration, either random or site specific, it would seem likely that an animal deficient in a major component of NHEJ would show less evidence of vector integration. The DNA-dependent protein kinase catalytic subunit is an integral component of NHEJ and is the locus of the underlying genetic defect in immunodeficient SCID mice. Song et al. (30) compared AAV integration both in vitro using cell extracts from wild-type C57BL/6 and SCID mice (derived from C57BL/6) and in vivo. The in vitro assay had been developed as an assay for site-specific integration. Interestingly, the presence of a DNA-dependent protein kinase catalytic subunit (DNAPKcs) in the wild-type extract appeared to inhibit AAV integration. The in vivo results were comparable. Hepatectomies were performed on animals 2 weeks after administration of AAV vectors carrying green fluorescent protein (GFP) as the transgene. Approximately 75% of the liver was removed, and regeneration was allowed to take place. In the SCID mice, a significant percentage (greater than 40%) of hepatocytes still expressed GFP, while GFP fluorescence was lower (less than 10%) in the regenerated wild-type livers. These results were interpreted to mean that either integration was more frequent in SCID mice and/or more stable; thus, DNAPKcs was inhibitory to persistent vector integration (which allowed transgene expression).In this paper, we report experiments designed to assess the effects of mutants which lead to defects in NHEJ on site-specific integration by AAV. We have also compared frequency and stability of site-specific integration relative to those of integration at other sites for both AAV containing single-stranded genomes and those with self-complementary genomes.  相似文献   

16.
17.
Adeno-associated virus type 2 is known to inhibit replication of herpes simplex virus 1 (HSV-1). This activity has been linked to the helicase- and DNA-binding domains of the Rep68/Rep78 proteins. Here, we show that Rep68 can bind to consensus Rep-binding sites on the HSV-1 genome and that the Rep helicase activity can inhibit replication of any DNA if binding is facilitated. Therefore, we hypothesize that inhibition of HSV-1 replication involves direct binding of Rep68/Rep78 to the HSV-1 genome.  相似文献   

18.
Baculovirus can transiently transduce primary human and rat hepatocytes, as well as a subset of stable cell lines. To prolong transgene expression, we have developed new hybrid vectors which associate key elements from adeno-associated virus (AAV) with the elevated transducing capacity of baculovirus. The hybrid vectors contain a transgene cassette composed of the β-galactosidase (β-Gal) reporter gene and the hygromycin resistance (Hygr) gene flanked by the AAV inverted terminal repeats (ITRs), which are necessary for AAV replication and integration in the host genome. Constructs were derived both with and without the AAV rep gene under the p5 and p19 promoters cloned in different positions with respect to the baculovirus polyheidrin promoter. A high-titer preparation of baculovirus-AAV (Bac-AAV) chimeric virus containing the ITR–Hygr–β-Gal sequence was obtained with insect cells only when the rep gene was placed in an antisense orientation to the polyheidrin promoter. Infection of 293 cells with Bac-AAV virus expressing the rep gene results in a 10- to 50-fold increase in the number of Hygr stable cell clones. Additionally, rep expression determined the localization of the transgene cassette in the aavs1 site in approximately 41% of cases as detected by both Southern blotting and fluorescent in situ hybridization analysis. Moreover, site-specific integration of the ITR-flanked DNA was also detected by PCR amplification of the ITR-aavs1 junction in transduced human fibroblasts. These data indicate that Bac-AAV hybrid vectors can allow permanent, nontoxic gene delivery of DNA constructs for ex vivo treatment of primary human cells.  相似文献   

19.
20.
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