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 765 毫秒
1.
2.
3.
Although precluded from using splicing to produce multiple small Rep proteins, adeno-associated virus type 5 (AAV5) generates a Rep40-like protein by alternative translation initiation at an internal AUG. A defined region upstream of the internal AUG was both required and sufficient to program internal initiation within AAV5 and may act similarly in heterologous contexts. The internally initiated AAV5 Rep40-like protein was functional and had helicase activity similar to that of AAV2 Rep40. Surprisingly, both the AAV5 Rep40-like protein and Rep52 were able to be translated from the AAV5 upstream P7-generated RNAs; however, the relative level of small to large Rep proteins was reduced compared to that of the wild type. A P19 mutant AAV5 infectious clone generated near-wild-type levels of the double-stranded monomer replicative form (mRF) replicative intermediate but reduced levels of virus, consistent with the previously defined role of Rep40-like proteins in genome encapsidation. Levels of mutant virus were dramatically reduced upon amplification.  相似文献   

4.
5.
6.
7.
8.
9.
Yoto Y  Qiu J  Pintel DJ 《Journal of virology》2006,80(3):1604-1609
Polyadenylation of B19 pre-mRNAs at the major internal site, (pA)p1, is programmed by the nonconsensus core cleavage and polyadenylation specificity factor-binding hexanucleotide AUUAAA. Efficient use of this element requires both downstream and upstream cis-acting elements and is further influenced by an adjacent AAUAAC motif. The primary hexanucleotide element must be nonconsensus to allow efficient readthrough of P6-generated pre-mRNAs into the capsid-coding region. An additional cleavage and polyadenylation site, (pA)p2, 296 nucleotides downstream of (pA)p1 was shown to be used following both B19 infection and transfection of a genomic clone. RNAs polyadenylated at (pA)p2 comprise approximately 10% of B19 RNAs that are polyadenylated internally.  相似文献   

10.
11.
12.
13.
Qiu J  Cheng F  Pintel D 《Journal of virology》2007,81(15):7974-7984
Adeno-associated virus type 5 (AAV5) is unique among human AAV serotypes in that it uses a polyadenylation site [(pA)p] within the single small intron in the center of the genome. We previously reported that inhibition of polyadenylation at (pA)p, necessary for read-through of P41-generated capsid gene pre-mRNAs which are subsequently spliced, requires binding of U1 snRNP to the upstream donor. Inhibition was reduced as the distance between the cap site and the donor was increased (increasing the size of the 5' exon). Here, we have demonstrated that U1-70K is a key component of U1 snRNP that mediates inhibition of polyadenylation at (pA)p. Furthermore, introduction of a U-rich stretch, predicted to target TIA-1 and thus increase the affinity of U1 snRNP binding to the intervening donor site, significantly augmented inhibition of (pA)p, while depletion of TIA-1 by siRNA increased (pA)p read-through. Finally, artificially tethering the cap binding complex (CBC) components CBP80 and CBP20 upstream of the intron donor increased inhibition of polyadenylation at (pA)p. Our results suggest that interaction with the CBC strengthens U1 snRNP binding to the downstream intron donor in a manner inversely proportional to the size of the 5' exon, thus governing the competition between intron splicing and polyadenylation at (pA)p. This competition must be optimized to program both the levels of polyadenylation of P7- and P19-generated RNA at (pA)p required to produce proper levels of the essential Rep proteins and the splicing of P41-generated RNAs to produce the proper ratio of capsid proteins during AAV5 infection.  相似文献   

14.
The DNA of human parvovirus adeno-associated virus type 2 (AAV) integrates preferentially into a defined region of human chromosome 19. Southern blots of genomic DNA from latently infected cell lines revealed that the provirus was not simply inserted into the cellular DNA. Both the proviral and adjoining cellular DNA organization indicated that integration occurred by a complex, coordinated process involving limited DNA replication and rearrangements. However, the mechanism for targeted integration has remained obscure. The two larger nonstructural proteins (Rep68 and Rep78) of AAV bind to a sequence element that is present in both the integration locus (P1) and the AAV inverted terminal repeat. This binding may be important for targeted integration. To investigate the mechanism of targeted integration, we tested the cloned integration site subfragment in a cell-free replication assay in the presence or absence of recombinant Rep proteins. Extensive, asymmetric replication of linear or open-circular template DNA was dependent on the presence of P1 sequence and Rep protein. The activities of Rep on the cloned P1 element are analogous to activities on the AAV inverted terminal repeat. Replication apparently initiates from a 3'-OH generated by the sequence-specific nicking activity of Rep. This results in a covalent attachment between Rep and the 5'-thymidine of the nick. The complexity of proviral structures can be explained by the participation of limited DNA replication facilitated by Rep during integration.  相似文献   

15.
Four Rep proteins are encoded by the human parvovirus adeno-associated virus type 2 (AAV). The two largest proteins, Rep68 and Rep78, have been shown in vitro to perform several activities related to AAV DNA replication. The Rep78 and Rep68 proteins are likely to be involved in the targeted integration of the AAV DNA into human chromosome 19, and the full characterization of these proteins is important for exploiting this phenomenon for the use of AAV as a vector for gene therapy. To obtain sufficient quantities for facilitating the characterization of the biochemical properties of the Rep proteins, the AAV rep open reading frame was cloned and expressed in Escherichia coli as a fusion protein with maltose-binding protein (MBP). Recombinant MBP-Rep68 and MBP-Rep78 proteins displayed the following activities reported for wild-type Rep proteins when assayed in vitro: (i) binding to the AAV inverted terminal repeat (ITR), (ii) helicase activity, (iii) site-specific (terminal resolution site) endonuclease activity, (iv) binding to a sequence within the integration locus for AAV DNA on human chromosome 19, and (v) stimulation of radiolabeling of DNA containing the AAV ITR in a cell extract. These five activities have been described for wild-type Rep produced from mammalian cell extracts. Furthermore, we recharacterized the sequence requirements for Rep binding to the ITR and found that only the A and A' regions are necessary, not the hairpin form of the ITR.  相似文献   

16.
Adeno-associated virus type 2 (AAV2) preferentially integrates its genome into the AAVS1 locus on human chromosome 19. Preferential integration requires the AAV2 Rep68 or Rep78 protein (Rep68/78), a Rep68/78 binding site (RBS), and a nicking site within AAVS1 and may also require an RBS within the virus genome. To obtain further information that might help to elucidate the mechanism and preferred substrate configurations of preferential integration, we amplified junctions between AAV2 DNA and AAVS1 from AAV2-infected HeLaJW cells and cells with defective Artemis or xeroderma pigmentosum group A genes. We sequenced 61 distinct junctions. The integration junction sequences show the three classical types of nonhomologous-end-joining joints: microhomology at junctions (57%), insertion of sequences that are not normally contiguous with either the AAV2 or the AAVS1 sequences at the junction (31%), and direct joining (11%). These junctions were spread over 750 bases and were all downstream of the Rep68/78 nicking site within AAVS1. Two-thirds of the junctions map to 350 bases of AAVS1 that are rich in polypyrimidine tracts on the nicked strand. The majority of AAV2 breakpoints were within the inverted terminal repeat (ITR) sequences, which contain RBSs. We never detected a complete ITR at a junction. Residual ITRs at junctions never contained more than one RBS, suggesting that the hairpin form, rather than the linear ITR, is the more frequent integration substrate. Our data are consistent with a model in which a cellular protein other than Artemis cleaves AAV2 hairpins to produce free ends for integration.  相似文献   

17.
In vitro replication of adeno-associated virus DNA.   总被引:23,自引:23,他引:0       下载免费PDF全文
The study of eukaryotic viral DNA replication in vitro has led to the identification of cellular enzymes involved in DNA replication. Adeno-associated virus (AAV) is distinct from previously reported systems in that it is believed to replicate entirely by leading-strand DNA synthesis and requires coinfection with adenovirus to establish completely permissive replication. In previous work, we demonstrated that two of the AAV nonstructural proteins, Rep78 and -68, are site-specific endonucleases and DNA helicases that are capable of resolving covalently closed AAV termini, a key step in AAV DNA replication. We have now cloned the AAV nonstructural proteins Rep78, Rep68, and Rep52 in the baculovirus expression system. Using the baculovirus-expressed proteins, we have developed an efficient in vitro AAV DNA replication system which mimics the in vivo behavior of AAV in every respect. With no-end AAV DNA as the starting substrate, the reaction required an adenovirus-infected cell extract and the presence of either Rep78 or Rep68. Rep52, as expected, did not support DNA replication. A mutant in the AAV terminal resolution site (trs) was defective for DNA replication in the in vitro assay. Little, if any, product was formed in the absence of the adenovirus-infected HeLa cell extract. In general, uninfected HeLa extracts were less efficient in supporting AAV DNA replication than adenovirus-infected extracts. Thus, the requirement for adenovirus infection in vivo was partially duplicated in vitro. The reduced ability of uninfected HeLa extracts to support complete DNA replication was not due to a defect in terminal resolution but rather to a defect in the reinitiation reaction or in elongation. Rep78 produced a characteristic monomer-dimer pattern of replicative intermediates, but surprisingly, Rep68 produced little, if any, dimer replicative form. The reaction had a significant lag (30 min) before incorporation of 32P-deoxynucleoside triphosphate could be detected in DpnI-resistant monomer replicative form and was linear for at least 4 h after the lag. The rate of incorporation in the reaction was comparable to that in the simian virus 40 in vitro system. Replication of the complete AAV DNA molecule was demonstrated by the following criteria. (i) Most of the monomer and dimer product DNAs were completely resistant to digestion with DpnI. (ii) Virtually all of the starting substrate was converted to heavy-light or heavy-heavy product DNA in the presence of bromo-dUTP when examined on CsCl density gradients.(ABSTRACT TRUNCATED AT 400 WORDS)  相似文献   

18.
The strand-specific, site-specific endonuclease (nicking) activity of the Rep68 and Rep78 (Rep68/78) proteins of adeno-associated virus type 2 (AAV) is involved in AAV replication, and appears to be involved in AAV site-specific integration. Rep68/78 cuts within the inverted terminal repeats (ITRs) of the AAV genome and in the AAV preferred integration locus on human chromosome 19 (AAVS1). The known endonuclease cut sites are 11-16 bases away from the primary binding sites, known as Rep recognition sequences (RRSs). A linear, double-stranded segment of DNA, containing an RRS and a cut site, has previously been shown to function as a substrate for the Rep68/78 endonuclease activity. We show here that mutation of the Rep recognition sequence, within such a DNA segment derived from the AAV ITRs, eliminates the ability of this substrate to be cleaved detectably by Rep78. Rep78 nicks the RRS-containing site from AAVS1 about half as well as the linear ITR sequence. Eighteen other RRS-containing sequences found in the human genome, but outside AAVS1, are not cleaved by Rep78. These results may help to explain the specificity of AAV integration.  相似文献   

19.
Adeno-associated virus type 2 (AAV) is the only known eucaryotic virus capable of targeted integration in human cells. AAV integrates preferentially into human chromosome (ch) 19q13.3qter. The nonstructural proteins of AAV-2, Rep78 and Rep68, are essential for targeted integration. Rep78 and Rep68 are multifunctional proteins with diverse biochemical activities, including site-specific binding to AAV and ch-19 target sequences, helicase activity, and strand-specific, site-specific endonuclease activities. Both a Rep DNA binding element (RBE) and a nicking site essential for AAV replication present within the viral terminal repeats are also located on ch-19. Recently, identical RBE sequences have been identified at other locations in the human genome. This fact raises numerous questions concerning AAV targeted integration; specifically, how many RBE sequences are in the human genome? How does Rep discriminate between these and the ch-19 RBE sequence? Does Rep interact with all sites and, if so, how is targeted integration within a fixed time frame facilitated? To better characterize the role of Rep in targeted integration, we established a Rep-dependent filter DNA binding assay using a highly purified Rep-68 fusion protein. Electron microscopy (EM) analysis was also performed to determine the characteristics of the Rep-RBE interaction. Our results determined that the Rep affinity for ch-19 is not distinct compared to other RBEs in the human genome when utilizing naked DNA. In fact, a minimum-binding site (GAGYGAGC) efficiently associated with Rep, suggesting that as many as 2 × 105 sites may exist. In addition, such sites also exist frequently in nonprimate mammalian genomes, although AAV integrates site specifically into primate genomes. EM analysis demonstrated that only one Rep-DNA complex was formed on ch-19 target DNA. Surprisingly, identically sized complexes were observed on all substrates containing a RBE sequence, but never on DNA lacking an RBE. Rep-DNA complexes involved a multimeric protein structure that spanned ca. 60 bp. Immunoprecipitation of AAV latently infected cells determined that 1,000 to 4,000 copies of Rep78 and Rep68 protein are expressed per cell. Comparison of the Rep association constant with those of established DNA binding proteins indicates that sufficient molecules of Rep are present to interact with all potential RBE sites. Moreover, Rep expression in the absence of AAV cis-acting substrate resulted in Rep-dependent amplification and rearrangement of the target sequence in ch-19. This result suggests that this locus is a hot spot for Rep-dependent recombination. Finally, we engineered mice to carry a single 2.7-kb human ch-19 insertion containing the AAV ch-19 target locus. Using cells derived from these mice, we demonstrated that this sequence was sufficient for site-specific recombination after infection with transducing vectors expressing Rep. This result indicates that any host factors required for targeting are conserved between human and mouse. Furthermore, the human ch-19 cis sequences and chromatin structure required for site-specific recombination are contained within this fragment. Overall, these results indicate that the specificity of targeted recombination to human ch-19 is not dictated by differential Rep affinities for RBE sites. Instead, specificity is likely dictated by human ch-19 sequences that serve as a Rep protein-mediated origin of replication, thus facilitating viral targeting through Rep-Rep interactions and host enzymes, resulting in site-specific recombination. Control of specificity is clearly dictated by the ch-19 sequences, since transfer of these sequences into the mouse genome are sufficient to achieve Rep-dependent site-specific integration.Adeno-associated virus type 2 (AAV) contains a single-stranded DNA genome of approximately 4.7 kb (50) and is a member of the Parvoviridae family (3). AAV is unique among other eucaryotic DNA viruses in that it utilizes a biphasic lifecycle to persist in nature. In the presence of a helper virus, adenovirus (Ad) or herpesvirus, AAV will undergo a productive infection. In the absence of a helper virus, AAV will integrate preferentially (>70%) into chromosome (ch) 19q13.3qter (3, 35). The ability of this nonpathogenic DNA virus, or virus-derived vector systems, to integrate site specifically have made it an attractive candidate vector for human gene therapy (45).The AAV genome consists of two open reading frames (ORFs), which comprise the rep and cap genes, and 145-bp inverted terminal repeats (ITRs), which serve as the origins of replication (3, 35). The left ORF of AAV encodes four nonstructural proteins, Rep78, Rep68, Rep52, and Rep40. Extensive characterization of Rep78 and Rep68 in vitro has identified the following biochemical activities, DNA binding (18, 19), site-specific and strand-specific endonuclease activities (17, 19), and DNA-RNA and DNA-DNA helicase activities (17, 19, 59), all of which appear to be necessary for viral replication (15, 53). More importantly, Rep78 and Rep68 are required for mediating targeted integration (2, 43, 47, 51, 60).Though site-specific integration is dependent upon either of the two large Rep proteins, the AAV ITRs are the only cis elements required for integration (34, 44, 61). In the absence of Rep proteins, the virus will still integrate through the ITR sequence but randomly into the host genome (21, 56, 61). Although integration in the absence of the Rep proteins is random, virus-cell junctions are nearly identical to junctions formed during targeted integration (DNA microhomology at junctions, specific deletions of the ITR sequences, rearrangement of the chromosome locus, and head-to-tail virus concatemers) (41, 62). In fact, in vitro integration products generated using cellular extracts produced identical type junctions, demonstrating the essential role the ITRs play in viral integration (62). From this analysis, Yang et al. (62) concluded that both random and targeted integration are dependent upon a cellular recombination pathway, with the role of Rep facilitating integration at ch-19. To help account for AAV targeting, a nearly identical Rep binding element (RBE) and a nicking site (trs) to that present on the AAV ITR was identified on the ch19.13.3qter AAV integration sequence (2325, 43, 46, 54, 57). It was also demonstrated that Rep68 could mediate complex formation between the AAV ITR and the ch-19 integration site in vitro (57). This led to a hypothesis that AAV may target integration by Rep-mediated complex formation between the AAV ITR and the ch19 integration site. However, since this observation subsequent data has demonstrated that Rep can bind to degenerate RBE sequences, (5, 32). In fact, computer analysis identified at least 15 genomic genes which contained RBE sites that bound to AAV Rep protein in vitro, all more efficient than the ch-19 sequence (58). These data raise the question as to how Rep can target ch-19 among other RBE sequences. Using an Epstein-Barr virus (EBV)-based shuttle vector system carrying sequences from ch-19, Linden et al. demonstrated that the trs site was also critical for AAV site-specific integration (29, 30). When the trs site was not present, targeting was lost, even though the RBE was present. The present study suggested that both sequences were essential for site-specific integration (the RBE and the trs sequences). The probability of identifying a RBE with the correct proximity of a trs site would suggest a frequency of <6 × 10−11/genome, thereby defining a unique sequence in the human genome (54). While these studies identify ch-19 cis elements required for AAV targeted integration and suggest why this reaction is specific, how Rep carries out this reaction remains unclear.Critical to any model of AAV Rep-mediated targeted integration is the ability to recognize the ch-19 target sequence among other potential RBE sequences. Though Rep can bind many degenerate sequences, the actual definition of what constitutes an RBE is somewhat unclear. Random oligonucleotide selection demonstrated that the RBE could be defined as an 8-bp sequence: 5′-GAGYGAGC-3′ (5). However, it was shown by methylation interference assays that the RBE was an 18-bp core sequence and that any mutation within this sequence would significantly affect Rep binding (42). Also, the report by Wonderling and Owens (58) demonstrated that the RBE oligonucleotides derived from the BLAST search contained mutations in this 18-bp core sequence but still bound better to the MBP-Rep68 than to the ch-19 RBE. Depending on the definition of an AAV RBE, the copy number present in the human genome (GAGYGAGC = 200,000 copies/genome, whereas 18-bp core = 1 copy/genome) could significantly impact the ability of Rep to identify its target locus.Based on the above information, the number of RBE sequences in the human genome, how Rep discriminates between these and the ch-19 target locus RBE sequence, and how Rep interacts with all sites and still facilitates targeted integration within a fixed time frame become of significant importance. In this study, we evaluated the role of alternative RBEs in the human genome and how these sequences might impact the ability of Rep to target the locus on ch-19. Using a filter-binding assay and a highly purified source of Rep68 protein, we established that genomic DNA will compete efficiently against a ch-19 target sequences. In this assay, a minimum Rep binding site of 8-bp in the context of large DNA fragments demonstrated competition, suggesting that as many as 200,000 potential binding sites may exist in the human genome. Filter-binding analysis of genomic DNA successfully retained ch-19 target sequences, as well as a cellular RBE identified by BLAST analysis, corroborating the competition results. Electron microscopy (EM) analysis was utilized to distinguish possible differences between Rep protein DNA interaction with ch-19 RBE compared to a minimum 8-bp RBE sequence. Identical multimeric Rep protein DNA complexes, which spanned about 60 bp, assembled on ch-19 target DNA, as well as a minimum RBE site, but never on heterologous DNA lacking these sequences. At a high Rep concentration, protein DNA looping structures were detected, but no evidence for paranemic structures were observed. In vivo analysis of Rep protein levels in a latent infection demonstrated approximately 1 to 4,000 copies/cell. Analysis of Rep expression in non-virus-infected cells demonstrated DNA rearrangement of the ch-19 target sequence, suggesting that this locus is a hot spot for Rep-induced DNA amplification and rearrangement that most likely influences AAV targeted integration. Finally, generation of an animal model carrying the human ch-19 sequence at the mouse hypoxanthine phosphoribosyltransferase (HPRT) locus facilitated AAV Rep-mediated targeted integration and corroborates the importance of the ch-19 RBE-trs sequence.  相似文献   

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