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.
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene, making it a potential target for gene therapy. There is, however, a scarcity of vectors that can accommodate the 14-kb DMD cDNA and permanently genetically correct muscle tissue in vivo or proliferating myogenic progenitors in vitro for use in autologous transplantation. Here, a dual high-capacity adenovirus-adeno-associated virus (hcAd/AAV) vector with two full-length human dystrophin-coding sequences flanked by AAV integration-enhancing elements is presented. These vectors are generated from input linear monomeric DNA molecules consisting of the Ad origin of replication and packaging signal followed by the recently identified AAV DNA integration efficiency element (p5IEE), the transgene(s) of interest, and the AAV inverted terminal repeat (ITR). After infection of producer cells with a helper Ad vector, the Ad DNA replication machinery, in concert with the AAV ITR-dependent dimerization, leads to the assembly of vector genomes with a tail-to-tail configuration that are efficiently amplified and packaged into Ad capsids. These dual hcAd/AAV hybrid vectors were used to express the dystrophin-coding sequence in rat cardiomyocytes in vitro and to restore dystrophin synthesis in the muscle tissues of mdx mice in vivo. Introduction into human cells of chimeric genomes, which contain a structure reminiscent of AAV proviral DNA, resulted in AAV Rep-dependent targeted DNA integration into the AAVS1 locus on chromosome 19. Dual hcAd/AAV hybrid vectors may thus be particularly useful to develop safe treatment modalities for diseases such as DMD that rely on efficient transfer and stable expression of large genes.  相似文献   

2.
Collaco RF  Cao X  Trempe JP 《Gene》1999,238(2):397-405
Adeno-associated virus (AAV) is a human parvovirus that is currently receiving widespread attention for its potential use as a gene therapy vector. Construction of the recombinant AAV vector (rAAV) involves replacing most of the viral genome with a transgene of interest and then packaging this recombinant genome into an infectious virion. Most current protocols for generating rAAV entail the co-transfection of a vector plasmid and a packaging plasmid that expresses the viral replication and structural genes onto adenovirus (Ad) infected cells growing in culture. Limitations of this procedure include (1) contamination of rAAV with the Ad helper virus, (2) low yields of rAAV and (3) production of replication-competent AAV. In this report we describe new helper plasmids (pSH3 and pSH5) that eliminate the Ad co-infection requirement. The helper plasmids express the AAV rep and cap genes and the Ad E2A, VAI and E4 genes. When the helper plasmids are co-transfected onto human 293 cells with a vector plasmid in the absence of Ad infection, the rAAV vector yield is up to 80-fold greater than those obtained with the pAAV/Ad packaging plasmid. Moreover, replication competent AAV in the rAAV preparations is less than 0.00125%. The major advantages of this system are (1) the absence of infectious adenovirus and (2) the use of only two plasmids, which enhances transfection efficiencies and hence vector production. We believe that this two-plasmid transfection system will allow for more widespread use of the AAV vector system because of its simplicity and high yields. This system will be especially useful for preclinical analyses of multiple rAAV vectors.  相似文献   

3.
Recently, we demonstrated that inverted repeat sequences inserted into first-generation adenovirus (Ad) vector genomes mediate precise genomic rearrangements resulting in vector genomes devoid of all viral genes that are efficiently packaged into functional Ad capsids. As a specific application of this finding, we generated adenovirus-adeno-associated virus (AAV) hybrid vectors, first-generation Ad vectors containing AAV inverted terminal repeat sequences (ITRs) flanking a reporter gene cassette inserted into the E1 region. We hypothesized that the AAV ITRs present within the hybrid vector genome could mediate the formation of rearranged vector genomes (DeltaAd.AAV) and stimulate transgene integration. We demonstrate here that DeltaAd.AAV vectors are efficiently generated as by-products of first-generation adenovirus-AAV vector amplification. DeltaAd.AAV genomes contain only the transgene flanked by AAV ITRs, Ad packaging signals, and Ad ITRs. DeltaAd.AAV vectors can be produced at a high titer and purity. In vitro transduction properties of these deleted hybrid vectors were evaluated in direct comparison with first-generation Ad and recombinant AAV vectors (rAAVs). The DeltaAd.AAV hybrid vector stably transduced cultured cells with efficiencies comparable to rAAV. Since cells transduced with DeltaAd.AAV did not express cytotoxic viral proteins, hybrid viruses could be applied at very high multiplicities of infection to increase transduction rates. Southern analysis and pulsed-field gel electrophoresis suggested that DeltaAd.AAV integrated randomly as head-to-tail tandems into the host cell genome. The presence of two intact AAV ITRs was crucial for the production of hybrid vectors and for transgene integration. DeltaAd.AAV vectors, which are straightforward in their production, represent a promising tool for stable gene transfer in vitro and in vivo.  相似文献   

4.
A prominent goal in gene therapy research concerns the development of gene transfer vehicles that can integrate exogenous DNA at specific chromosomal loci to prevent insertional oncogenesis and provide for long-term transgene expression. Adenovirus (Ad) vectors arguably represent the most efficient delivery systems of episomal DNA into eukaryotic cell nuclei. The most advanced recombinant Ads lack all adenoviral genes. This renders these so-called high-capacity (hc) Ad vectors less cytotoxic/immunogenic than those only deleted in early regions and creates space for the insertion of large/multiple transgenes. The versatility of hcAd vectors is been increased by capsid modifications to alter their tropism and by the incorporation into their genomes of sequences promoting chromosomal insertion of exogenous DNA. Adeno-associated virus (AAV) can insert its genome into a specific human locus designated AAVS1. Trans- and cis-acting elements needed for this reaction are the AAV Rep78/68 proteins and Rep78/68-binding sequences, respectively. Here, we describe the generation, characterization and testing of fiber-modified dual hcAd/AAV hybrid vectors (dHVs) containing both these elements. Due to the inhibitory effects of Rep78/68 on Ad-dependent DNA replication, we deployed a recombinase-inducible gene switch to repress Rep68 synthesis during vector rescue and propagation. Flow cytometric analyses revealed that rep68-positive dHVs can be produced similarly well as rep68-negative control vectors. Western blot experiments and immunofluorescence microscopy analyses demonstrated transfer of recombinase-dependent rep68 genes into target cells. Studies in HeLa cells and in the dystrophin-deficient myoblasts from a Duchenne muscular dystrophy (DMD) patient showed that induction of Rep68 synthesis in cells transduced with fiber-modified and rep68-positive dHVs leads to increased stable transduction levels and AAVS1-targeted integration of vector DNA. These results warrant further investigation especially considering the paucity of vector systems allowing permanent phenotypic correction of patient-own cell types with large DNA (e.g. recombinant full-length DMD genes).  相似文献   

5.
The adeno-associated virus (AAV) vector system is based on nonpathogenic and helper-virus-dependent parvoviruses. The vector system offers safe, efficient, and long-term in vivo gene transfer in numerous tissues. Clinical trials using AAV vectors have demonstrated vector safety as well as efficiency. The increasing interest in the use of AAV for clinical studies demands large quantities of vectors and hence a need for improvement in vector production. The commonly used transient-transfection method, although versatile and free of adenovirus (Ad), is not cost-effective for large-scale production. While the wild-type-Ad-dependent AAV producer cell lines seem to be cost-effective, this method faces the problem of wild-type Ad contamination. To overcome these shortcomings, we have explored the feasibility of creating inducible AAV packaging cell lines that require neither transfection nor helper virus infection. As a first step toward that goal, we have created a cell line containing highly inducible Ad E1A and E1B genes, which are essential for AAV production. Subsequently, the AAV Rep and Cap genes and an AAV vector containing a green fluorescent protein (GFP) reporter gene were stably introduced into the E1A-E1B cell line, generating inducible AAV-GFP packaging cell lines. Upon induction of E1A and E1B genes and infection with replication-defective Ad with E1A, E1B, and E3 deleted, the packaging cells yielded high-titer AAV-GFP vectors. Finally, the E2, E4, and VA genes of Ad, under the control of their endogenous promoters, were also introduced into these cells. A few producer cell lines were obtained, which could produce AAV-GFP vectors upon simple drug induction. Although future improvement is necessary to increase the stability and vector yield of the cells, our study has nonetheless demonstrated the feasibility of generating helper-virus-free inducible AAV producer cell lines.  相似文献   

6.

Background

Helper‐dependent, or gutted, adenoviruses (Ad) lack viral coding sequences, resulting in reduced immunotoxicity compared with conventional Ad vectors. Gutted Ad growth requires a conventional Ad to supply replication and packaging functions in trans. Methods that allow high‐titer growth of gutted vectors while reducing helper contamination, and which use safer helper viruses, will facilitate the use of gutted Ad vectors in vivo.

Methods

Replication‐defective helper viruses were generated that are deleted for Ad E1, E2b and E3 genes, but which contain loxP sites flanking the packaging signal. Complementing Ad packaging cell lines (C7‐cre cells) were also generated by transfecting 293 cells with the Ad E2b genes encoding DNA polymerase and pre‐terminal protein, and with a cre‐recombinase plasmid.

Results

We show that C7‐cre cells allow efficient production of gutted Ad using ΔE1 + ΔE2b + ΔE3 helper viruses whose growth can be limited by cre‐loxP‐mediated excision of the packaging signal. Gutted Ad vectors carrying ~28 kb cassettes expressing full‐length dystrophin were prepared at high titers, similar to those obtained with E2b+ helpers, with a resulting helper contamination of <1%.

Conclusions

These new packaging cell lines and helper viruses offer several significant advantages for gutted Ad vector production. They allow gutted virus amplification using a reduced number of passages, which should reduce the chances of selecting rearranged products. Furthermore, the residual helper contamination in gutted vector preparations should be less able to elicit immunological reactions upon delivery to tissues, since E2b‐deleted vectors display a profound reduction in viral gene expression. Copyright © 2002 John Wiley & Sons, Ltd.
  相似文献   

7.
Smith RH  Afione SA  Kotin RM 《BioTechniques》2002,33(1):204-6, 208, 210-1
Adeno-associated viruses (AAVs) are replication-defective parvoviruses that require helper virusfunctionsfor efficient productive replication. The AAVs are currently premier candidates as vectors for human gene therapy applications. In particular; much recent interest has been expressed concerning recombinant AAV serotype 5 (rAAV-5) vectors, as they appear to utilize cellular receptors distinctfrom those of the prototypical AAV serotype (AAV-2) and have been reported to have transduction properties in vivo that differ significantly from those of the prototype. One of the most popular current methodsfor the production of rAAVs involves co-transfection of human 293 cells with three plasmids: (i) an adenovirus (Ad)-derived helper plasmid containing Ad genes required for AAV replication, (ii) an AAV-derived plasmid encoding complementing AAV genes (ie., the viral rep and cap genes), and (iii) a target plasmid containing a transgene of interestflanked by AAV inverted terminal repeats (ITRs) that confer packaging and replication capabilities upon the ITR-flanked heterologous DNA. Here we describe novel plasmid reagents designed for convenient and efficient production of rAAV-S. An integrated helper plasmid containing all Ad genes requiredfor the efficient production of recombinant AAV as well as the complementing AAV genes on the same plasmid backbone, was constructed via transposase-mediated insertion into an Ad helper plasmid of a transposable element containing the AAV-5 rep and cap genes linked to a selectable marker This simple strategy can be used in the rapid and efficient construction of integrated helper plasmids derived from any reported AAV serotype for which a molecular clone exists.  相似文献   

8.
As vectors, adenoviruses (Ads) have many attractive advantages for in vivo gene therapy. However, Ads do not usually integrate into the host genome and gene expression is, thus, transient. Adeno-associated virus (AAV) integrates into a specific locus (AAVS1) on the human host's chromosome 19, while conventional recombinant AAV (rAAV) vectors do not possess this property because such vectors lack the rep gene. AAV vectors carrying the rep gene do not have enough space for insertion of a transgene. We have constructed a hybrid adenovirus/adeno-associated virus (Ad/AAV) vector which has the advantages of both Ads and AAVs. Given that the rep gene products inhibit propagation of Ads, we used the Cre/loxP-expression-switching system to regulate the expression of the rep gene. The Ad/AAV vector easily propagates, can efficiently infect a broad range of cell types, and can integrate into a specific locus on host chromosomes.  相似文献   

9.
10.
Helper-dependent (HD) adenovirus vectors devoid of all viral coding sequences have a large cloning capacity and provide long-term transgene expression in vivo with negligible toxicity, making them attractive vectors for gene therapy. Currently, the most efficient means of producing HD vectors involves coinfecting 293 cells expressing Cre with the HD vector and a helper virus bearing a packaging signal flanked by loxP sites. Cre-mediated packaging signal excision renders the helper virus genome unpackageable but still able to replicate and provide helper functions for HD vector propagation. Typically, helper virus contamination is < or =1% pre- and < or =0.1% postpurification by CsCl banding. While these contamination levels are low, further reduction is desirable. However, this objective has not been realized since the Cre/loxP system was first developed. This lack of progress is due, at least in part, to our lack of understanding of the origins of the contaminating helper virus, thus rendering its reduction or elimination difficult to achieve. This study was designed to investigate the possible sources of contaminating helper virus persisting during HD vector amplification. The results revealed that Cre is limiting in helper virus-infected Cre-expressing 293 cells, thereby permitting helper viruses to escape packaging signal excision and propagate. The results of this study should provide a foundation for developing rational strategies to further reduce or possibly eliminate the contaminating helper virus.  相似文献   

11.
The serotypes of adeno-associated virus (AAV) have the potential to become important resources for clinical gene therapy. In an effort to compare the role of serotype-specific virion shells on vector transduction, we cloned each of the serotype capsid coding domains into a common vector backbone containing AAV type 2 replication genes. This strategy allowed the packaging of AAV2 inverted terminal repeat vectors into each serotype-specific virions. Each of these helper plasmids (pXR1 through pXR5) efficiently replicated the transgene DNA and expressed helper proteins at nearly equivalent levels. In this study, we observed a correlation between the amount of transgene replication and packaging efficiency. The physical titer of these hybrid vectors ranged between 1.3 x 10(11) and 9.8 x 10(12)/ml (types 1 and 2, respectively). Of the five serotype vectors, only types 2 and 3 were efficiently purified by heparin-Sepharose column chromatography, illustrating the high degree of similarity between these virions. We analyzed vector transduction in reference and mutant Chinese hamster ovary cells deficient in heparan sulfate proteoglycan and saw a correlation between transduction and heparan sulfate binding data. In this analysis, types 1 and 5 were most consistent in transduction efficiency across all cell lines tested. In vivo each serotype was ranked after comparison of transgene levels by using different routes of injection and strains of rodents. Overall, in this analysis, type 1 was superior for efficient transduction of liver and muscle, followed in order by types 5, 3, 2, and 4. Surprisingly, this order changed when vector was introduced into rat retina. Types 5 and 4 were most efficient, followed by type 1. These data established a hierarchy for efficient serotype-specific vector transduction depending on the target tissue. These data also strongly support the need for extending these analyses to additional animal models and human tissue. The development of these helper plasmids should facilitate direct comparisons of serotypes, as well as begin the standardization of production for further clinical development.  相似文献   

12.
Mini-adenoviruses (mAd) deleted of all viral coding regions represent an emerging approach for transgene expression. We have exploited the unique features of the adeno-associated virus (AAV) terminal repeats within the context of an adenovirus-adeno-associated hybrid virus (Ad/AAV) as a strategy for rapid and efficient generation of mAd. Excision and generation of mAd from the parental Ad/AAV hybrid vector was achieved in 293 cells through recombination but without selection for mAd production. Analysis of mAd isolated from 293 cells indicated that mAd DNA exists as monomer and dimer forms within the recombinant viral capsid. Formation of recombinant mAd was significantly increased using an AAV Rep78- or Rep68-expressing cell line through Rep-mediated excision utilizing the AAV terminal repeat sequences present in the Ad/AAV hybrid virus genome. The mAd viruses were infectious and able to transfer functional gene to A549 and HeLa cells. This approach is rapid and efficient, thereby providing a simplified methodology for generating mAd with functional transducing capabilities.  相似文献   

13.
14.
Cao L  Liu Y  During MJ  Xiao W 《Journal of virology》2000,74(24):11456-11463
Recombinant adeno-associated virus (rAAV) is capable of directing long-term, high-level transgene expression without destructive cell-mediated immune responses. However, traditional packaging methods for rAAV vectors are generally inefficient and contaminated with replication-competent AAV (rcAAV) particles. Although wild-type AAV is not associated with any known human diseases, contaminating rcAAV particles may affect rAAV gene expression and are an uncontrolled variable in many AAV gene transfer studies. In the current study, a novel strategy was designed to both optimize AAV rep gene expression and increase vector yield, as well as simultaneously to diminish the potential of generating rcAAV particles from the helper plasmid. The strategy is based on the insertion of an additional intron in the AAV genome. In the AAV infectious clone, the intron insertion had no effects on the properties of Rep proteins expressed. Normal levels of both Rep and Cap proteins were expressed, and the replication of the AAV genome was not impaired. However, the generation of infectious rcAAV particles using intronized AAV helper was greatly diminished, which was due to the oversized AAV genome caused by the insertion of the artificial introns. Moreover, the rAAV packaging was significantly improved with the appropriate choice of intron and insertion position. The intron is another element that can regulate the rep and cap gene expression from the helper plasmid. This study provides for a novel AAV packaging system which is highly versatile and efficient. It can not only be combined with other AAV packaging systems, including rep-containing cell lines and herpes simplex virus hybrid packaging methods, but also be used in other vector systems as well.  相似文献   

15.
Herpes simplex virus type 1 (HSV-1)-based amplicon vectors have a large transgene capacity and can efficiently infect many different cell types. One disadvantage of HSV-1 vectors is their instability of transgene expression. By contrast, vectors based on adeno-associated virus (AAV) can either persist in an episomal form or integrate into the host cell genome, thereby supporting long-term gene expression. AAV expresses four rep genes, rep68, -78, -40, and -52. Of those, rep68 or rep78 are sufficient to mediate site-specific integration of the AAV DNA into the host cell genome. The major disadvantage of AAV vectors is the small transgene capacity ( approximately 4.6 kb). In this study, we constructed HSV/AAV hybrid vectors that contained, in addition to the standard HSV-1 amplicon elements, AAV rep68, rep78, both rep68 and -78, or all four rep genes and a reporter gene that was flanked by the AAV inverted terminal repeats (ITRs). Southern blots of Hirt DNA from cells transfected with the hybrid vectors and HSV-1 helper DNA demonstrated that both the AAV elements and the HSV-1 elements were functional in the context of the hybrid vector. All hybrid vectors could be packaged into HSV-1 virions, although those containing rep sequences had lower titers than vectors that did not. Site-specific integration at AAVS1 on human chromosome 19 was directly demonstrated by PCR and sequence analysis of ITR-AAVS1 junctions in hybrid vector-transduced 293 cells. Cell clones that stably expressed the transgene for at least 12 months could easily be isolated without chemical selection. In the majority of these clones, the transgene cassette was integrated at AAVS1, and no sequences outside the ITR cassette, rep in particular, were present as determined by PCR, ITR rescue/replication assays, and Southern analysis. Some of the clones contained random integrations of the transgene cassette alone or together with sequences outside the ITR cassette. These data indicate that the long-term transgene expression observed following transduction with HSV/AAV hybrid vectors is, at least in part, supported by chromosomal integration of the transgene cassette, both randomly and site specifically.  相似文献   

16.
Bernt K  Liang M  Ye X  Ni S  Li ZY  Ye SL  Hu F  Lieber A 《Journal of virology》2002,76(21):10994-11002
We have developed a new class of adenovirus vectors that selectively replicate in tumor cells. The vector design is based on our recent observation that a variety of human tumor cell lines support DNA replication of adenovirus vectors with deletions of the E1A and E1B genes, whereas primary human cells or mouse liver cells in vivo do not. On the basis of this tumor-selective replication, we developed an adenovirus system that utilizes homologous recombination between inverted repeats to mediate precise rearrangements within the viral genome resulting in replication-dependent activation of transgene expression in tumors (Ad.IR vectors). Here, we used this system to achieve tumor-specific expression of adenoviral wild-type E1A in order to enhance viral DNA replication and spread within tumor metastases. In vitro DNA replication and cytotoxicity studies demonstrated that the mechanism of E1A-enhanced replication of Ad.IR-E1A vectors is efficiently and specifically activated in tumor cells, but not in nontransformed human cells. Systemic application of the Ad.IR-E1A vector into animals with liver metastases achieved transgene expression exclusively in tumors. The number of transgene-expressing tumor cells within metastases increased over time, indicating viral spread. Furthermore, the Ad.IR-E1A vector demonstrated antitumor efficacy in subcutaneous and metastatic models. These new Ad.IR-E1A vectors combine elements that allow for tumor-specific transgene expression, efficient viral replication, and spread in liver metastases after systemic vector application.  相似文献   

17.
Herpes simplex virus type 1 (HSV-1) amplicon vectors are promising gene delivery tools, but their utility in gene therapy has been impeded to some extent by their inability to achieve stable transgene expression. In this study, we examined the possibility of improving transduction stability in cultured human cells via site-specific genomic integration mediated by adeno-associated virus (AAV) Rep and inverted terminal repeats (ITRs). A rep(-) HSV/AAV hybrid amplicon vector was made by inserting a transgene cassette flanked with AAV ITRs into an HSV-1 amplicon backbone, and a rep(+) HSV/AAV hybrid amplicon was made by inserting rep68/78 outside the rep(-) vector 3' AAV ITR sequence. Both vectors also had a pair of loxP sites flanking the ITRs. The resulting hybrid amplicon vectors were successfully packaged and compared to a standard amplicon vector for stable transduction frequency (STF) in human 293 and Gli36 cell lines and primary myoblasts. The rep(+), but not the rep(-), hybrid vector improved STF in all three types of cells; 84% of Gli36 and 40% of 293 stable clones transduced by the rep(+) hybrid vector integrated the transgene into the AAVS1 site. Due to the difficulty in expanding primary myoblasts, we did not assess site-specific integration in these cells. A strategy to attempt further improvement of STF by "deconcatenating" the hybrid amplicon DNA via Cre-loxP recombination was tested, but it did not increase STF. These data demonstrate that introducing the integrating elements of AAV into HSV-1 amplicon vectors can significantly improve their ability to achieve stable gene transduction by conferring the AAV-like capability of site-specific genomic integration in dividing cells.  相似文献   

18.
To achieve stable gene transfer into human hematopoietic cells, we constructed a new vector, DeltaAd5/35.AAV. This vector has a chimeric capsid containing adenovirus type 35 fibers, which conferred efficient infection of human hematopoietic cells. The DeltaAd5/35.AAV vector genome is deleted for all viral genes, allowing for infection without virus-associated toxicity. To generate high-capacity DeltaAd5/35.AAV vectors, we employed a new technique based on recombination between two first-generation adenovirus vectors. The resultant vector genome contained an 11.6-kb expression cassette including the human gamma-globin gene and the HS2 and HS3 elements of the beta-globin locus control region. The expression cassette was flanked by adeno-associated virus (AAV) inverted terminal repeats (ITRs). Infection with DeltaAd5/35.AAV allowed for stable transgene expression in a hematopoietic cell line after integration into the host genome through the AAV ITR(s). This new vector exhibits advantages over existing integrating vectors, including an increased insert capacity and tropism for hematopoietic cells. It has the potential for stable ex vivo transduction of hematopoietic stem cells in order to treat sickle cell disease.  相似文献   

19.
Viral vectors are emerging as potent basic research tools and gene therapy vehicles in many laboratory animal models. However, little information is available on the potential shedding of these vectors and the consequent exposure risk to investigators and animal care staff from animals over time. This study provides empirical information to Institutional Biosafety Committees and animal care programs, to enhance their ability to perform risk management of laboratory animals treated with viral vectors. Control experiments evaluated the limit of detection of third-generation lentivirus, recombinant adeno-associated virus, and E1-deleted adenovirus tested directly from stocks and after application onto cage plastic or bedding. After inoculation of ICR or NOD-SCID mice, we quantified the recovery of viral vector genomes directly from blood, urine, and fecal samples and assessed the persistence of infectious vector at the site of injection and from soiled bedding at different time points after inoculation. No differences were seen between ICR and NOD-SCID mice. We saw no evidence of vector amplification after in vivo inoculation. The most environmentally persistent vector was recombinant adeno-associated virus, which has no known pathogenicity in humans. In light of these data, we conclude that commonly used replication-deficient viral vectors pose minimal exposure risk by 72 h after inoculation. Prudent precautions at Animal Biosafety Level 2 are warranted during initial administration, but Level 1 safety measures may be sufficient after cage changing and biosafety evaluation.Abbreviations: Ad, adenovirus; CMV, cytomegalovirus; EGFP, enhanced green fluorescent protein; LV, third-generation lentivirus; qPCR, qualitative PCR; rAAV, recombinant adeno-associated virusThe era of customizable transgenic mice and molecular manipulation of viruses to deliver targeted genes has revolutionized our ability to study gene-specific functions. A typical viral vector is engineered by separating the wildtype virus genome onto multiple nonoverlapping plasmids, with the minimal coding sequences necessary for replication provided in trans to the transgene construct, which contains only those cis-acting viral elements necessary to direct packaging into a noninfectious virion. Large portions of the genome necessary for pathogenicity are completely removed. The resulting engineered viral vectors offer the ability to specifically target tissues and genes and are emerging as potent gene therapy vehicles. With these tools for basic research, we are equipped to answer questions about the effects of genes and proteins on development, behavior, sensation, and organ function and in disease.Each viral vector has unique properties and targets defined cell populations (Figure 1).1,18 For example, lentiviral (LV) vectors are used for in vivo gene delivery because they efficiently transduce both dividing and nondividing cells and stably integrate into the host genome, providing long-term transgene expression. Lentiviral vectors typically are based on HIV1 that has been substantially debilitated to provide multiple safeguards against the production of replication-competent lentivirus.6,32 Third-generation lentiviral vectors have deletions in the promoter region of the long terminal repeats, rendering these vectors self-inactivating after proviral integration. In addition, they include deletions in all 6 major genes involved in pathogenesis—including the genes for Env, Vpr, Vpu, Vif, Nef, Rev, and (in some versions) Tat. Typically, vesicular stomatitis virus glycoprotein is substituted for the native viral Env protein. The transient packaging system contains less than 30% of the original viral genome, and the produced replication-deficient self-inactivating vector particles integrate less than 8% of the HIV genome into infected cells. Such an HIV1-based vector, pseudotyped with the vesicular stomatitis virus glycoprotein as its major envelope protein, is the most commonly used lentiviral vector and is available commercially from most major life-science vendors and from core facilities at various scientific institutions.Open in a separate windowFigure 1.Viral vector characteristics.Adenoviral vectors are small (90 nm) nonenveloped icosahedral vectors with a linear double-stranded DNA genome. They infect dividing and nondividing cells but do not actively integrate into the host cell genome; therefore, expression is transient in actively dividing cells and tissues. Long-term transgene expression in nondividing cells and tissues has been achieved as well. Adenoviral vectors are most commonly derived from adenoviral serotype 5 (Ad5).9 Helper-free, replication-defective recombinant Ad5 vectors often are generated through deletion of the essential E1a/b and nonessential E3 regions of the viral genome. Removal of these sequences allows the introduction of a gene of interest into the deleted region, with a packaging capacity of approximately 8 kb. Adenoviral replication is dependent on the E1a/b region of the viral genome; as such, recombinant Ad5 vectors are unable to replicate, and packaging of replication-defective Ad5 vector particles is achieved through the transfection of a linearized plasmid containing the recombinant Ad5 vector genome into HEK293/17 cells, or derivatives of this cell line, which stably express the Ad5 E1a gene. With E1a/b- and E3-deleted Ad5 vectors, 2 separate recombination events would need to occur during packaging to generate replication-competent wildtype virus. ‘Gutless’ (that is, helper-dependent) Ad5 viral vector systems have also been developed; in these systems, all viral genes have been removed from the recombinant vector genome. This manipulation greatly increases the packaging capacity of these vectors—from approximately 8 kb to 36 kb—and markedly reduces immune responses in vivo. Helper virus is required for the production of gutless adenoviral vector, and various strategies have been developed to remove this helper virus from the subsequent viral vector preparation.25Recombinant adeno-associated viral (rAAV) vectors are derived from the AAV2 virus, a very small (20 nm) icosahedral nonenveloped virus with a linear single-stranded DNA genome that does not actively integrate into the host cell genome. Wildtype AAV2 is not a known human pathogen, and coinfection with a helper virus, such as adenovirus, is required for AAV2 to replicate within a host cell. All of the wildtype viral genome has been deleted from rAAV2 vectors, except for the 5′ and 3′ inverted terminal repeat regions, which are the only cis acting, noncoding viral sequences necessary for packaging of a recombinant vector genome into replication-defective particles. rAAV2 vectors typically are produced in a helper-virus-free system.34 The recombinant rAAV2 genome expressing the gene of interest, adenoviral helper genes, and the AAV coding sequences for the rep and cap proteins are provided on separate plasmids and are transiently transfected into producer cells, such as HEK293T/17. The packaging capacity of rAAV2 vectors is approximately 4.7 kb, although larger genomes have successfully been packaged or expressed in vivo. The produced, replication-defective rAAV2 vector particles can also be cross-packaged with capsid proteins from other AAV serotypes22 as well as engineered capsid proteins,15,31,33 offering attractive systems for altering the cell and tissue tropism of the produced vector.Institutional Biosafety Committees serve a critical function in providing oversight and balanced risk assessment of hazard use in research facilities, effectively managing the risk associated with viral vector use, both in the laboratory and in vivo after treatment of laboratory animals. Decisions related to the biosafety of viral vector use often are based on the known pathogenic properties of the parental wildtype viruses, in combination with case-by-case determination of additional risks related to the characteristics of the particular viral vector subtype and the biologic activity (if known) of the expressed transgene or regulatory nucleic acid to be packaged. Similar strategies are used to further classify these agents into 1 of 4 specific risk groups.3 Replication-deficient Ad and LV vectors currently are classified in section III-D-3-a as group 2 viruses, and experiments with these agents involving animals are categorized into section III-D-4-b.20 Although AAV vectors are classified as group 1 viruses, the biologic activity of the expressed transgene may lead an Institutional Biosafety Committee to mandate the use of Biosafety Level 2 or Animal Biosafety Level 2 precautions with rAAV vectors. To date, few published reports address direct measurement of the exposure risk from animals treated with different viral vectors have been available to Institutional Biosafety Committee members. Given this paucity of information, recommendations frequently are made based on the biologic properties of the parent (often virulent and replicative) strains of these vectors. These biologic properties most specifically relate to virulence, pathogenicity, infectious dose, environmental stability, route of spread, and communicability of the wildtype strain.30Several potential risks are associated with these agents, including transmission of the viral vector itself, homologous recombination resulting in the generation of replication-competent vector or virulent wildtype-like virus, contamination by residual helper viruses (if used during vector preparation), and adverse effects of the inserted gene or regulatory nucleic acid (for example, oncogenesis).17,27 The wildtype, parental HIV and adenovirus strains can be stable for a month or longer in a laboratory setting.29 Although important in the context of traditional virus research, these guidelines are only broadly applicable to advanced, later-generation agents that have been specifically and rationally modified to deliver target genes without generating associated vector-induced disease or uncontrolled replication.Managing these vector agents in rodents according to Animal Biosafety Level 2 guidelines can sometimes be problematic when the use of primary engineering controls (for example, a biologic safety cabinet) is impractical or impossible. The ability to incorporate suggested safety measures may be limited, in part, because of the experimental design, infrastructural limitation, and work processes. Examples of these procedures include behavioral testing, breeding, bioimaging, MRI and CT scanning, and other specialized research activities that require the use of large or immobile equipment. In addition, routine practices in vivaria may increase the potential for contamination thorough direct contact with infected animals, fomite contamination, or potential aerosolization through handling of soiled bedding.A group of investigators at our institution are developing and using novel or modified viral vectors in various unconventional ways. These methodologies are expanding rapidly to other disciplines, and many institutions are witnessing the expansion of ‘gray areas’ in risk assessment and containment. Guidance in evaluating the safety and use of LV vectors is available,19 but few reports detailing empirical data are available to support these concepts. If the real threat of shedding or infection can be quantified, it then would be feasible to establish accurately the risk associated with treated animals and enhance laboratory and vivarium safety practices.We hypothesized that risks associated with viral vectors in mice may be lower than those generally associated with the parent viruses. Once given to animals, the current state-of-the-art replication-deficient viral gene therapy and targeting vectors likely pose minimal risk of exposure to staff, and application of standard Animal Biosafety Level 2 biohazard precautions when working with rodents carrying viral vectors may not be necessary indefinitely. We report here our evaluation of the risk of the shedding of several classes of genetically modified viral vectors from rodents and subsequent potential for human exposure to these vectors.  相似文献   

20.
Direct or inverse repeated sequences are important functional features of prokaryotic and eukaryotic genomes. Considering the unique mechanism, involving single-stranded genomic intermediates, by which adenovirus (Ad) replicates its genome, we investigated whether repetitive homologous sequences inserted into E1-deleted adenoviral vectors would affect replication of viral DNA. In these studies we found that inverted repeats (IRs) inserted into the E1 region could mediate predictable genomic rearrangements, resulting in vector genomes devoid of all viral genes. These genomes (termed DeltaAd.IR) contained only the transgene cassette flanked on both sides by precisely duplicated IRs, Ad packaging signals, and Ad inverted terminal repeat sequences. Generation of DeltaAd.IR genomes could also be achieved by coinfecting two viruses, each providing one inverse homology element. The formation of DeltaAd.IR genomes required Ad DNA replication and appeared to involve recombination between the homologous inverted sequences. The formation of DeltaAd. IR genomes did not depend on the sequence within or adjacent to the inverted repeat elements. The small DeltaAd.IR vector genomes were efficiently packaged into functional Ad particles. All functions for DeltaAd.IR replication and packaging were provided by the full-length genome amplified in the same cell. DeltaAd.IR vectors were produced at a yield of approximately 10(4) particles per cell, which could be separated from virions with full-length genomes based on their lighter buoyant density. DeltaAd.IR vectors infected cultured cells with the same efficiency as first-generation vectors; however, transgene expression was only transient due to the instability of deleted genomes within transduced cells. The finding that IRs present within Ad vector genomes can mediate precise genetic rearrangements has important implications for the development of new vectors for gene therapy approaches.  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