Effect of Genome Size on AAV Vector Packaging (2024)

  • Journal List
  • Mol Ther
  • v.18(1); 2010 Jan
  • PMC2839202

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Effect of Genome Size on AAV Vector Packaging (1)

Molecular Therapy

Mol Ther. 2010 Jan; 18(1): 80–86.

Published online 2009 Nov 10. doi:10.1038/mt.2009.255

PMCID: PMC2839202

PMID: 19904234

Zhijian Wu,1 Hongyan Yang,1 and Peter Colosi1

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

Adeno-associated virus (AAV) vector genomes have been limited to 5 kilobases (kb) in length because their packaging limit was thought to be similar to the size of the parent AAV genome. Recent reports claim that significantly larger vector genomes can be packaged intact. We examined the packaged vector genomes from plasmid-encoded AAV vectors that ranged from 4.7 to 8.7kb in length, using AAV types 2, 5, and 8 capsids. Southern blot analysis indicated that packaged AAV vector genomes never exceeded 5.2kb in length irrespective of the size of the plasmid-encoded vector or the capsid type. This result was confirmed by vector genome probing with strand-specific oligonucleotides. The packaged vector genomes derived from plasmid-encoded vectors exceeding 5kb were heterogeneous in length and truncated on the 5′ end. Despite their truncated genomes, vector preparations produced from plasmid-encoded vectors exceeding 5.2kb mediated reporter gene expression in vitro at high multiplicity of infection (MOI). The efficiency of expression was substantially lower than that of reporter vectors with genomes <5kb in length. We propose that transcriptionally functional, intact vector genomes are generated in cells transduced at high MOI from the fragmentary genomes of these larger vectors, probably by recombination.

Introduction

The adeno-associated viruses (AAVs) are single-stranded DNA viruses with genomes that are ~4.7 kilobases (kb) in length. They comprise the Dependovirus branch of the parvovirus family and require co-infection with a helper virus, typically adenovirus or herpes virus, to replicate. Recombinant vectors derived from AAVs are attractive for the development of human therapeutics because they mediate long-term gene expression and the parent AAVs produce no known pathology in humans. AAV vectors were recently used to successfully treat patients with Leber congenital amaurosis, a severe form of genetically acquired childhood blindness.1,2,3 A substantial limitation of AAV vectors is their small packaging capacity that is generally considered to be <5kb. This limitation makes vector design for diseases involving larger genes challenging or impossible. Larger genes are relatively common and are involved in diseases such as cystic fibrosis, duch*enne muscular dystrophy, hemophilia A, and some forms of Leber congenital amaurosis and retinitis pigmentosa. To address this problem, strategies based on trans-splicing4,5,6 and hom*ologous recombination7 were developed. All of these methods require multiple vectors, and the efficiency of some of these methods is inherently limited by the efficiencies of intron processing or hom*ologous recombination. Two recent reports suggest that AAV capsids are capable of packaging substantially larger vector genomes in an intact form. Grieger and Samulski8 reported that AAV types 1–5 can package up to 6kb DNA. Most recently, Allocca et al.9 reported that AAV type 5 can package up to 8.9kb DNA and mediate efficient transgene expression both in vitro and in vivo.

In this study, we evaluated the packaged genome size and in vitro expression characteristics of AAV vectors ranging from 4.7 to 8.7kb in length. The vectors examined were a mouse CEP290 vector and a set of lacZ reporter vectors of increasing length. These were packaged into AAV types 2, 5, and 8 capsids using the three-plasmid transfection method. We found that the size of packaged vector genome DNAs never exceeded 5.2kb no matter what size vector was encoded by the production plasmid. This was true for all capsid types and vectors tested. The packaged vector genome DNA from plasmid-encoded vectors exceeding 5.2kb consisted of a heterogeneous population of genomes with a broad size distribution and a maximum length of ~5kb. The heterogeneity was generated by deletion on the 5′ end. Vector preparations consisting of heterogeneous fragmentary vector genomes were capable of gene expression in vitro when used at high multiplicity of infection (MOI). The expression efficiency of these vector preparations was substantially lower than vector preparations with genomes that were <5kb in length.

Results

AAV 2, 5, and 8 capsids do not efficiently package an intact 8.6kb AAV mCEP290 vector genome

Mutations in the human CEP290 gene account for ~15% of patients with Leber congenital amaurosis,10 a severe form of inherited blindness. CEP290 is a ubiquitously expressed, 2,480 amino-acid protein that plays roles in ciliary transport and centrosomal function.11 The rd16 mouse carries an in-frame deletion in the CEP290 gene and exhibits an early-onset retinal degeneration that is similar to the human disease.11 In an attempt to produce a vector that could be used to complement the CEP290 deficiency in the rd16 mouse, we constructed an AAV vector plasmid that encoded a mouse CEP290 expression cassette driven by the cytomegalovirus (CMV) promoter (Figure 1a). The total length of this AAV vector genome was 8.6kb including the two inverted terminal repeats (ITRs). We attempted to package this vector into AAV types 2, 5, and 8 capsids using a conventional three-plasmid transfection method.12 Vector yields, as measured by quantitative PCR, were not markedly different between the three AAV serotypes but were approximately tenfold lower than yields from vectors with lengths <4,850 base pairs (bp). Vector genome DNA was purified from the vector preparations, and subjected to alkaline electrophoresis and Southern analysis to assess the size and integrity of the packaged vector genomes (Figure 1b). In contrast to previous studies,8,9,13 our results showed that the packaged vector genomes consisted of a heterogeneous population of genomes with a broad size distribution and a maximum length of ~5kb. No intact 8.6kb vector genomes were detected. Fragmentary genome packaging was observed with all three serotypes. AAV5 displayed no particular ability to package large vector genomes.

Open in a separate window

Figure 1

Electrophoresis of AAV-mCEP290 vectors. (a) Structure of the AAV mouse CEP290 vector. (b) Alkaline agarose electrophoresis and Southern blot analysis of isolated AAV mCEP290 vector genomes. Lanes 1 and 2: 8,343bp vector plasmid DNA marker, 0.1 and 1ng, respectively; lanes 3 and 4: 5,067bp vector plasmid DNA marker, 0.1 and 1ng, respectively; lanes 5–7: DNA from 1 × 1010 vector genomes isolated from AAV2 mCEP290, AAV5 mCEP290, and AAV8 mCEP290 vector preparations, respectively. AAV, adeno-associated virus; bp, base pair; CMV, cytomegalovirus; ITR, inverted terminal repeat; kb, kilobase.

The packaged AAV 5 mCEP290 vector genomes are truncated on the 5′ end

To further characterize the packaged AAV5 mCEP290 vector genome population, strand-specific oligonucleotides corresponding to positions located throughout the mCEP290 vector were used to probe dilutions of dot-blotted vector genomes and control plasmid DNA that encodes the complete vector (Figure 2a). The strength of the hybridization signal from the packaged genome was compared to the signal from a linearized vector plasmid control (Figure 2b). The hybridization signals for control plasmid DNA were strong for all probes irrespective of hybridization position in the vector sequence. For the isolated vector genome DNA, the strongest hybridization signals were observed for probes that hybridize near the 3′ end of the vector genome. Almost no vector sequences occurring >5.2kb from the 3′ end of the vector were detected. This was true for probes specific to either DNA strand. This result indicated that the majority of the vector genomes in the population have intact 3′ ends but truncated 5′ ends. The truncations occurred at multiple positions located <5.2kb from the 3′ end of the vector genome. Our study did not determine whether the 5′ ends of the packaged genomes contain an ITR.

Open in a separate window

Figure 2

Genome integrity of AAV5-mCEP290 vector. (a) Hybridization positions of single-stranded oligonucleotides on the vector genome. (b) Strand-specific dot-blot hybridization of vector genome DNA. Twofold serial dilutions of vector plasmid DNA (top row) and isolated AAV5 mCEP290 vector genome DNA (bottom row). The masses of the vector plasmid DNAs are 50, 25, 12.5, 6.25, 3.1, 1.6, 0.78, and 0.39ng (wells 1–8, top row). The calculated masses of the isolated vector DNAs are 7, 3.5, 1.7, 0.86, 0.43, 0.21, 0.11, and 0.05ng (wells 1–8, bottom row). The DNA in the first well on the bottom row was isolated from 2.3 × 109 vector genomes.

Estimation of the size limit for efficient packaging of intact vector genomes

To estimate the packaging limit for AAV 2 and 5 capsids, a set of AAV lacZ vectors of increasing length were constructed (Figure 3a). The size of the plasmid-encoded vectors ranged from 4.7 to 8.7kb. All of these vectors contained a 4.4kb CMV lacZ expression cassette, located adjacent to the ITR or centrally, and phage lambda DNA was used to increase the total length of the vectors. After packaging the vectors, alkaline gel electrophoresis and Southern analysis were carried out on purified vector genome DNAs to assess their size (Figure 3b). The results were similar for vectors packaged in AAV2 and AAV5 capsids. Both the 4.7 and 5.2kb vectors showed clear bands at their respective sizes. Vector genome DNAs derived from plasmid-encoded vectors larger than 5.2kb consisted of a heterogeneous population of genomes with a broad size distribution and a maximum length of ~5kb. The upper packaging limit for both AAV2 and AAV5 capsids appears to be ~5.2kb.

Open in a separate window

Packaging of AAV2 and AAV5 lacZ vectors. (a) Structures of plasmid-encoded vectors ranging from 4.7 to 8.7kb. All of these vectors contained a 4.4kb CMV lacZ expression cassette, located adjacent to the ITR (I designation in name) or centrally (C designation in name), and phage lambda DNA was used to increase the total length. (b) Southern blot analysis of isolated vector genome DNA separated on an alkaline agarose gel. 1 × 1010 vector genomes were analyzed for the AAV5 vectors (upper panel), and 1 × 109 vector genomes were analyzed for the AAV2 vectors (lower panel). Vector plasmid DNA markers in lanes M1: 8.6kb (1ng), M2: 5.1kb (1ng), M3: 8.6kb (0.1ng), and M4 5.1kb (0.1ng). The DNA in M2 is an unintended partial digestion. Lanes 1–8 in both panels: genomes isolated from AAV-lacZ-4.7k, AAV-lacZ-I5.2k, AAV-lacZ-I5.7k, AAV-lacZ-I6.7k, AAV-lacZ-I8.7k, AAV-lacZ-C5.7k, AAV-lacZ-C6.7k, and AAV-lacZ-C8.7k vector preparations, respectively. CMV, cytomegalovirus; ITR, inverted terminal repeat; kb, kilobase.

Effect of vector size on transgene expression efficiency in vitro

The AAV type 2 vector preparations made from plasmid-encoded vectors ranging from 4.7 to 8.7kb in length (Figure 3a) were assessed for expression in vitro. Subconfluent 293 cells were infected with these preparations at MOIs of 2 × 103 and 1 × 104 vector genomes per cell. After 24 hours, the cultures were fixed, stained with X-gal, and counted (Figure 4a). The expression efficiency of the vectors fell into three groups. The vector preparation made from the 4.7kb plasmid-encoded vector (pAAV-lacZ-4.7k) had the highest expression at both MOIs and alone comprised the first group (Figure 4b). The second group was made up of vector preparations made from plasmid-encoded vectors that were longer than 5kb in length but that had intact lacZ expression cassettes and one ITR located entirely within 5,050bp of continuous DNA. This group was 4- to 15-fold less efficient than AAV2-lacZ-4.7k at both MOIs and included AAV2-lacZ-I5.2k, AAV2-lacZ-I5.7k, AAV2-lacZ-I6.7k, AAV2-lacZ-I8.7k, and AAV2-lacZ-C5.7k. The third group was made up of vector preparations made from plasmid-encoded vectors that were longer than 5kb in length but did not have intact lacZ expression cassettes and one ITR located entirely within 5,050bp of continuous DNA. This group had expression efficiencies ranging from 77- to 1,600-fold lower than AAV2-lacZ-4.7k depending on the MOI and included AAV2-lacZ-C6.7k and AAV2-lacZ-C8.7k. Most of the vector preparations showed a two- to fourfold increase in transduction efficiency when the MOI was increased fivefold. The expression efficiency of AAV2-lacZ-C8.7k preparation was extremely sensitive to the fivefold increase in MOI and increased 18-fold.

Open in a separate window

Figure 4

LacZ expression in 293 cells infected with AAV2-lacZ vectors. (a) LacZ expression in 293 cells infected with AAV2-lacZ vectors at MOI 2,000 (left panels) or 10,000 (right panels). (b) Quantification of transduction of the AAV2 lacZ vector set. MOI, multiplicity of infection.

Interestingly, all of the vector preparations mediated some degree of expression even though Southern analysis could not detect packaged genomes >5.2kb in length. It appears that vectors that can package at least one ITR and the entire lacZ expression cassette in ≤5,050bp of continuous DNA have greater expression efficiencies than those that cannot.

Discussion

Several groups have examined AAV packaging and have tried to assess the maximum packaging capacity of AAV vectors. Dong et al.14 were the first to systematically address the subject and reported that AAV type 2 vectors made from plasmid-encoded vectors exceeding 4.9kb showed substantially reduced expression when used to transduce cells in vitro. Subsequently, Grieger and Samulski8 reported that genomes up to 6.0kb could be packaged in AAV types 1–5 capsids, but that vectors exceeding 5.2kb were largely degraded in infected cells by the proteasome. Two years later, Wu et al.13 reported that a self-complementary vector with an intact 6.6kb genome could be produced using the AAV type 2 capsid. Most recently, Allocca et al.9 reported that AAV type 5 was able to package intact AAV genomes up to 8.9kb in length and that these vector preparations mediated gene expression in vitro and in vivo.

In this study, we investigated ability of AAV types 2, 5, and 8 capsids to package nine different plasmid-encoded AAV vectors. The vectors examined were an 8.6kb mouse CEP290 vector and a set of lacZ reporter vectors that ranged from 4.7 to 8.7kb in length. In contrast to some of the previous studies, we found that maximum size of the packaged vector DNA never exceeded ~5.2kb, no matter what size plasmid-encoded vector or AAV serotype was used. Given the limit of sensitivity of our Southern analysis (0.33% of the applied vector genome DNA), if intact 8.6kb CEP290 or 8.7kb lacZ genomes were packaged, they comprised <1 part in 300 (by mass) of the total packaged DNA. Additionally, we found that vector genomes isolated from vector preparations produced with plasmid-encoded vectors exceeding 5.2kb were heterogeneous with a broad size distribution and had a maximum length of ~5kb. Strand-specific probing of packaged CEP290 vector genomes indicated they were exclusively derived from the 5kb region located at the 3′ end of the vector genomes (of both polarities). Apparently, the genomes were truncated at the 5′ end. This may be due to the fact that AAV vectors package genomes unidirectionally into preformed capsids starting with the 3′ end.15 With larger vectors, the packaging process may stall after the capsid is full. The remaining unpackaged 5′ end of the genome may then be removed by cellular nucleases.

We then asked whether these fragmentary vector genomes, packaged in AAV type 2 capsids, were capable of mediating gene expression in cells. Somewhat surprisingly, all of the AAV2 lacZ vector preparations mediated some degree of reporter gene expression when used to transduce 293 cells at MOIs exceeding 1. The reason that vector preparations with fragmentary genomes mediate gene expression is not clear. A likely possibility is that overlapping vector genomes of opposite polarities anneal and regenerate full-length vector genomes in cells that are infected by multiple vector particles (Figure 5). Generation of full-length vector genomes may also occur by hom*ologous recombination. Multiple infections are probably common at the high MOIs used in our experiment. Intracellular vector recombination of AAV vectors has been previously described.7

Open in a separate window

Figure 5

Proposed model for packaging and transduction of AAV2 lacZ vectors. (1) Packaging, (2) uncoating, (3) second-strand DNA synthesis, (4) annealing, and (5) hom*ologous recombination.

The location of the 4.4kb CMV lacZ cassette in the vector genome had a large effect on transduction activity of the vectors. Vector preparations produced from 5.2 to 8.7kb plasmid-encoded vectors with intact lacZ expression cassettes and one ITR located entirely within 5,050bp of continuous DNA-mediated expression are about tenfold more efficiently than vector preparations derived from plasmid-encoded vectors that did not have intact lacZ expression cassettes and one ITR located entirely within 5,050bp of continuous DNA. The higher efficiency of vectors with ITR-proximal lacZ expression cassettes may indicate that these vectors simply package the first 5,050bp from the 3′ end, which encodes one ITR and an intact lacZ expression, and that this partial genome is capable of mediating gene expression without having to anneal to a second vector. This idea is illustrated in Figure 6.

Open in a separate window

Figure 6

Proposed model for fragmentary genome transduction. Right panel: one ITR and intact lacZ cassette contained in DNA fragment <5,050bp in length. Left panel: intact lacZ cassette and either ITR contained in a DNA fragment >5,050bp in length. The higher transduction efficiency of vectors with ITR-proximal lacZ expression cassettes may indicate that these vectors package the first 5,050bp from the 3′ end, which encodes one ITR and an intact lacZ expression, and that this fragmentary genome is capable of mediating gene expression without having to anneal to a second vector. bp, base pair; CMV, cytomegalovirus; ITR, inverted terminal repeat.

The transduction efficiency of the AAV2-lacZ-C8.7k vector preparation alone demonstrated a very high sensitivity to MOI, jumping 18-fold, versus two- to fourfold for the other vectors, in response to a fivefold increase in MOI. This vector and the AAV2-lacZ-C6.7k vector do not have intact lacZ expression cassettes within 5.2kb of either end of their genomes and would likely require intracellular recombination to regenerate an intact lacZ expression cassette. Assuming a maximum packaged genome size of 5.2kb, the AAV2-lacZ-C8.7k vector would have the smallest overlap between genomes of opposite polarities (1.7kb versus 3.7kb for the AAV2-lacZ-C6.7k vector). This smaller overlap may have made the expression efficiency of this vector more MOI sensitive. The genome populations in these vector preparations are heterogeneous with respect to size. Certainly, the smaller overlap would reduce the fraction of genomes capable of annealing within the cell.

It is also unclear why our data do not agree with previous reports that vector genomes as large as 8.9kb can be packaged and are functional for transduction. To control for differences in packaging plasmids and vector purification techniques, we repeated our experiments with two other sets of packaging plasmids9,16 and one other purification protocol17 with no change in outcome (data not shown).

In conclusion, we found no evidence that AAV capsids efficiently package vector genomes that are substantially larger than the wild-type viral genomes. The limitations imposed by the packaging limits of AAV capsids are major hindrances to the development of many otherwise promising and clinically relevant applications. If AAV vector-based gene delivery is to be used for these applications, strategies need to be developed to accommodate the genome size limitation. The recent reports of large genome packaging usually come with steep reductions in packaging or transduction efficiency. Considering our data, or the data of Dong et al.,14 this is not surprising. More work certainly needs to be done to see whether capsid or genome modifications can be found that will allow the efficient packaging of larger genomes. But we believe that the current trend of simply trying to package larger genomes into the commonly used AAV capsids is akin to fitting a small shoe onto a large foot.

Materials and Methods

AAV vector constructs and production. AAV2 ITRs were used in all AAV vector constructs. The mouse CEP290 complementary DNA was provided by Anand Swaroop (National Eye Institute/National Institutes of Health). To clone the mCEP290 complementary DNA into an AAV vector plasmid, PCR was carried out using the following primers: 5′-AGT CAT CGA TGC CAC CAT GCC ACC TAA TAT AAA GTG GAA AGA ATT AA-3′ and 5′-AGT CAG ATC TCT AAT AAA TAG GGA AAC TAT-3′. The PCR product was digested with Cla I and Bgl II, and then inserted into an AAV vector plasmid with an expression cassette consisting of a CMV promoter and a human β-globin polyadenylation site. The parent plasmid was described previously and was used for enhanced green fluorescent protein expression.18 The resulting plasmid was designated pV4.6c mCEP290. The plasmid encoding the 4.7kb, CMV lacZ vector was previously described.19 Phage lambda DNA of different lengths was used to create the larger lacZ vectors. PCR was used to create lambda DNA fragments of the appropriate sizes. The PCR products were inserted with either 3′ or 5′ of the CMV lacZ expression cassette. The designations of the resulting vectors are shown in Figure 3.

AAV production and purification was carried out as previously described.12 Real-time PCR was employed to titer the vectors. The following fluorescence-labeled probe (5′-TCC AAA ATG TCG TAA CAA CT-3′) and primers (5′-TGG GAG TTT GTT TTG CAC CAA-3′ and 5′-CGC CTA CCG CCC ATT TG-3′) were used for all vectors.

Alkaline gel electrophoresis and Southern blot analysis of AAV vector DNA. DNA was extracted from 1 × 109 or 1 × 1010 full vector particles as previously described.9 Before extraction, the vector particles were treated with DNase I to remove unpackaged DNA. Alkaline agarose gel electrophoreses was performed as previously described.20 DIG-labeled (Roche, Indianapolis, IN) or 32P-labeled probes were employed for DNA hybridization. To analyze AAV mCEP290 vectors, a 408bp Eco RI fragment on pV4.6c mCEP290 was used as a probe. The 5.1 and 8.3kb markers were produced by digestion of the pV4.6c mCEP290 with Sac II and Sph I, or Not I alone. To analyze AAV LacZ vectors, a 657bp Spe I-Age I fragment from the vector plasmid was used as a probe. The 5.1 and 8.6kb markers were generated by Sma I digestion of pAAV-lacZ-I5.2k and pAAV-lacZ-C8.7k plasmids, respectively.

Strand-specific dot-blot hybridization of AAV5 mCEP290 vector. AAV5 mCEP290 vector DNA was extracted as previously described.9 Extracted vector DNA from 2.3 × 109 vector genomes was applied to the first well of the manifold. Twofold serial dilutions of the extracted vector DNA were applied to the remaining seven wells of the manifold. The corresponding amounts of vector DNA applied to the eight wells of the manifold were 7, 3.5, 1.7, 0.86, 0.43, 0.21, 0.11, and 0.05ng. Standard DNA generated by Sma I digestion of pV4.6c mCEP290 was diluted in the same fashion. The amounts of plasmid DNA dotted onto the eight wells of the manifold were 50, 25, 12.5, 6.25, 3.1, 1.6, 0.78, and 0.39ng. Both vector and plasmid DNA were denatured by 0.25mol/l NaOH before loading to nylon membrane. Single-strand oligonucleotides corresponding to several positions in the vector genome (Table 1) were designed, synthesized, and labeled using DIG Oligonucleotide 3′-End Labeling Kit (Roche). Hybridization was carried out overnight at 10°C below the melting temperature of each probe. After probing, the membrane was stripped of DIG-labeled probe by washing twice with 0.2mol/l NaOH/0.1% sodium dodecyl sulfate for 15 minutes at 37°C and rinsing in 2× saline-sodium citrate for 5 minutes.

Table 1

Probes for strand-specific hybridization of AAV5-mCEP290 genome

Open in a separate window

Infection and quantification of transduced cells. HEK 293 cells were seeded on 24-well plates at a concentration of 1 × 105 cells per well 40 hours before infection. The cells were transfected with pAdeno5, plasmid encoding the E2A, VA RNA, and E4 regions of adenovirus type 2 (ref. 19). The cells were then infected with the AAV2 LacZ vectors at MOIs of 2 × 103 or 1 × 104. Etoposide was added to each well to produce a final concentration of 20nmol/l. After 24 hours, the cells were fixed with phosphate-buffered saline containing 2% formaldehyde and 0.2% glutaraldehyde for 5 minutes at room temperature. After fixation, lacZ expression was evaluated by histochemical staining with a solution of 1mg/ml X-gal in phosphate-buffered saline containing 5mmol/l K3Fe(CN)6, 5mmol/l K4Fe(CN)63H2O, and 2mmol/l MgCl2. The cells were stained for 4 hours at 37°C. Eight fields per well were counted, and the average number of blue cells per well was determined.

Acknowledgments

We thank Chun Y Gao of the National Eye Institute for assistance with cell imaging.

REFERENCES

  • Maguire AM, Simonelli F, Pierce EA, Pugh EN Jr, Mingozzi F, Bennicelli J, et al. Safety and efficacy of gene transfer for Leber's congenital amaurosis. N Engl J Med. 2008;358:2240–2248. [PMC free article] [PubMed] [Google Scholar]
  • Bainbridge JW, Smith AJ, Barker SS, Robbie S, Henderson R, Balaggan K, et al. Effect of gene therapy on visual function in Leber's congenital amaurosis. N Engl J Med. 2008;358:2231–2239. [PubMed] [Google Scholar]
  • Cideciyan AV, Aleman TS, Boye SL, Schwartz SB, Kaushal S, Roman AJ, et al. Human gene therapy for RPE65 isomerase deficiency activates the retinoid cycle of vision but with slow rod kinetics. Proc Natl Acad Sci USA. 2008;105:15112–15117. [PMC free article] [PubMed] [Google Scholar]
  • Nakai H, Storm TA., and , Kay MA. Increasing the size of rAAV-mediated expression cassettes in vivo by intermolecular joining of two complementary vectors. Nat Biotechnol. 2000;18:527–532. [PubMed] [Google Scholar]
  • Yan Z, Zhang Y, Duan D., and , Engelhardt JF. Trans-splicing vectors expand the utility of adeno-associated virus for gene therapy. Proc Natl Acad Sci USA. 2000;97:6716–6721. [PMC free article] [PubMed] [Google Scholar]
  • Sun L, Li J., and , Xiao X. Overcoming adeno-associated virus vector size limitation through viral DNA heterodimerization. Nat Med. 2000;6:599–602. [PubMed] [Google Scholar]
  • Halbert CL, Allen JM., and , Miller AD. Efficient mouse airway transduction following recombination between AAV vectors carrying parts of a larger gene. Nat Biotechnol. 2002;20:697–701. [PubMed] [Google Scholar]
  • Grieger JC., and , Samulski RJ. Packaging capacity of adeno-associated virus serotypes: impact of larger genomes on infectivity and postentry steps. J Virol. 2005;79:9933–9944. [PMC free article] [PubMed] [Google Scholar]
  • Allocca M, Doria M, Petrillo M, Colella P, Garcia-Hoyos M, Gibbs D, et al. Serotype-dependent packaging of large genes in adeno-associated viral vectors results in effective gene delivery in mice. J Clin Invest. 2008;118:1955–1964. [PMC free article] [PubMed] [Google Scholar]
  • den Hollander AI, Roepman R, Koenekoop RK., and , Cremers FP. Leber congenital amaurosis: genes, proteins and disease mechanisms. Prog Retin Eye Res. 2008;27:391–419. [PubMed] [Google Scholar]
  • Chang B, Khanna H, Hawes N, Jimeno D, He S, Lillo C, et al. In-frame deletion in a novel centrosomal/ciliary protein CEP290/NPHP6 perturbs its interaction with RPGR and results in early-onset retinal degeneration in the rd16 mouse. Hum Mol Genet. 2006;15:1847–1857. [PMC free article] [PubMed] [Google Scholar]
  • Grimm D, Zhou S, Nakai H, Thomas CE, Storm TA, Fuess S, et al. Preclinical in vivo evaluation of pseudotyped adeno-associated virus vectors for liver gene therapy. Blood. 2003;102:2412–2419. [PubMed] [Google Scholar]
  • Wu J, Zhao W, Zhong L, Han Z, Li B, Ma W, et al. Self-complementary recombinant adeno-associated viral vectors: packaging capacity and the role of rep proteins in vector purity. Hum Gene Ther. 2007;18:171–182. [PubMed] [Google Scholar]
  • Dong JY, Fan PD., and , Frizzell RA. Quantitative analysis of the packaging capacity of recombinant adeno-associated virus. Hum Gene Ther. 1996;7:2101–2112. [PubMed] [Google Scholar]
  • King JA, Dubielzig R, Grimm D., and , Kleinschmidt JA. DNA helicase-mediated packaging of adeno-associated virus type 2 genomes into preformed capsids. EMBO J. 2001;20:3282–3291. [PMC free article] [PubMed] [Google Scholar]
  • Rabinowitz JE, Rolling F, Li C, Conrath H, Xiao W, Xiao X, et al. Cross-packaging of a single adeno-associated virus (AAV) type 2 vector genome into multiple AAV serotypes enables transduction with broad specificity. J Virol. 2002;76:791–801. [PMC free article] [PubMed] [Google Scholar]
  • Wu Z, Sun J, Zhang T, Yin C, Yin F, Van Dyke T, et al. Optimization of self-complementary AAV vectors for liver-directed expression results in sustained correction of hemophilia B at low vector dose. Mol Ther. 2008;16:280–289. [PubMed] [Google Scholar]
  • Park TK, Wu Z, Kjellstrom S, Zeng Y, Bush RA, Sieving PA, et al. Intravitreal delivery of AAV8 retinoschisin results in cell type-specific gene expression and retinal rescue in the Rs1-KO mouse. Gene Ther. 2009;16:916–926. [PMC free article] [PubMed] [Google Scholar]
  • Matsush*ta T, Elliger S, Elliger C, Podsakoff G, Villarreal L, Kurtzman GJ, et al. Adeno-associated virus vectors can be efficiently produced without helper virus. Gene Ther. 1998;5:938–945. [PubMed] [Google Scholar]
  • Sambrook J., and , Russell DW.eds) (2001Molecular Cloning: A Laboratory Manual Cold Spring Harbor Laboratory Press: Cold Spring Harbor, New York. pp 999 [Google Scholar]

Articles from Molecular Therapy are provided here courtesy of The American Society of Gene & Cell Therapy

Effect of Genome Size on AAV Vector Packaging (2024)
Top Articles
Latest Posts
Article information

Author: Corie Satterfield

Last Updated:

Views: 6482

Rating: 4.1 / 5 (62 voted)

Reviews: 85% of readers found this page helpful

Author information

Name: Corie Satterfield

Birthday: 1992-08-19

Address: 850 Benjamin Bridge, Dickinsonchester, CO 68572-0542

Phone: +26813599986666

Job: Sales Manager

Hobby: Table tennis, Soapmaking, Flower arranging, amateur radio, Rock climbing, scrapbook, Horseback riding

Introduction: My name is Corie Satterfield, I am a fancy, perfect, spotless, quaint, fantastic, funny, lucky person who loves writing and wants to share my knowledge and understanding with you.