Abstract
One of the major challenges in the treatment of primary muscle disorders, which often affect many muscle groups, is achieving efficient, widespread transgene expression in muscle. In utero gene transfer can potentially address this problem by accomplishing gene delivery when the tissue mass is small and the immune system is immature. Previous studies with systemic in utero adeno-associated viral (AAV) vector serotype 1 gene delivery to embryonic day 16 (E-16) pups resulted in high levels of transduction in diaphragm and intercostal muscles, but no detectable transgene expression in limb muscles. Recently newer AAV serotypes such as AAV8 have demonstrated widespread and high transgene expression in skeletal muscles and diaphragm by systemic delivery in adult and neonatal mice. We tested AAV8 vector gene delivery by intraperitoneal administration in E-16 mice in utero. Using an AAV8 vector carrying a lacZ reporter gene, we observed high level transduction of diaphragm and intercostal muscles and more moderate transduction of multiple limb muscles and heart. Our current studies demonstrate the potential of AAV8 to achieve widespread muscle transduction in utero and suggest its therapeutic potential for primary muscle disorders.
Introduction
Several genetically-determined muscle disorders are caused by loss-of-function mutations and present at birth or in early childhood. Viral vectors delivered in utero offer a potential means of gene therapy for these disorders that primarily affect striated muscle.
Adeno-associated viral (AAV) vectors provide efficient gene delivery to striated muscle tissues.1 Recently identified AAV serotypes have varied tissue tropism and transduction efficiency. Among the currently known serotypes, AAV8 demonstrates superior potential to deliver transgenes to muscle tissues.2–9 Using systemic delivery of AAV8 vectors, reporter gene and therapeutic gene transfer studies in postnatal mice and hamsters demonstrated efficient transduction of skeletal muscle and heart in neonates and adults.6,7 Similarly Nakai et al. demonstrated the ability of AAV8 vector to deliver a transgene to skeletal muscle by systemic gene delivery in adult mice.6 Other studies also showed AAV8 vector effectively transduced nonhuman primate skeletal muscles.5
In disease models, AAV8 vectors have demonstrated potential for therapeutic benefit. In response to AAV8 vector-mediated myostatin-inhibitor gene transfer to mdx mice, skeletal muscle mass increased.4 AAV8 vector-mediated α-glucosidase gene transfer decreased glycogen storage in skeletal muscle of a mouse model of Pompe disease.3,9
Furthermore, the AAV8 vector appeared to be less immunogenic10 and demonstrated more rapid uncoating post-internalization11 compared to the AAV2 vector. In addition, the AAV8 vector also demonstrated good potential to deliver transgenes in various animal models such as mouse, rat and non-human primates.7,12–14 The AAV8 vector also showed high level transduction in various tissues such as pancreatic cells15, neuronal cells16,17, liver cells18, and smooth muscle cells.17
Although AAV8 vectors have been extensively studied in various animal models and in different age groups, their transduction potential in utero remains unknown. Preclinical studies have shown that in utero gene delivery results in good expression of transgene extending into adulthood. Intramuscular injection of AAV119 and AAV220 vectors into fetal muscle demonstrated good gene expression. However, systemic delivery of AAV1 vector in utero did not transduce limb muscle tissue to any significant degree.19 This study therefore aims to determine the distribution of AAV8 vector particles to different tissues and levels of expression after in utero systemic delivery.
Materials and methods
Production of AAV8lacZ vector
AAV8 vector carrying a lacZ expression cassette driven by the human cytomegalovirus (HCMV) promoter (AAV8lacZ) vector stocks were generated by the triple-plasmid transfection method.21 Briefly, the three plasmids are the AAV-CMV-lacZ vector plasmid, the mini-adeno helper plasmid, and the AAV8 packaging plasmid containing the AAV2 Rep gene and AAV8 Cap gene, which has an ATG to ACG start codon mutation to increase the vector yield.22 AAV8lacZ viral particles were purified by double CsCl gradient centrifugation and dialyzed three times against PBS containing 5% Sorbitol. The titer of vector genomes was determined by a standard DNA dot-blot assay.
Mice and in utero AAV8lacZ vector administration
Timed pregnant CD1 mice (Harlan Sprangue Dawley Inc, Maryland) at embryonic day 16 (E-16) were used for these studies according to the protocol approved by the University of Pittsburgh Institutional Animal Care and Use Committee. The pregnant mice were anesthetized with an (intraperioneal) IP injection of 5mg/kg of xylazine and 50mg/kg of ketamine. After shaving the abdominal wall and cleaning the skin with betadine, a single vertical incision was made through the abdominal wall and peritoneal membrane to expose the uterus. Fetal injections were performed under sterile conditions and high magnification. AAV8lacZ was injected intraperitoneally. A fluorescent marker (fluorescent beads or fluorescent dextran) was also injected as a means to identify treated mice after birth. A total volume of 8–10 μl of AAV8lacZ solution/fetus was injected using a 33 G needle (Hamilton, USA) attached to a PB600 syringe dispenser (Hamilton, USA). A dose of 6.4 × 1011 vector genomes were injected per fetus. For this study, we injected 14 pups from 9 pregnant CD1 mothers to achieve 8 positive pups. A total of 8 treated and 8 untreated littermate pups were studied.
Between 2 and 5 days following injection, the naturally delivered pups were examined using a fluorescence microscope to identify those pups that were injected by the presence of the fluorescent marker. At 9 weeks following injection, different tissues from the mother, the injected pups and uninjected littermates were analyzed for biodistribution of β-galactosidase (β-gal) expression and vector content.
Whole muscle staining
Tissues such as forelimb muscle, hindlimb muscle, heart, diaphragm, intercostal muscle and abdominal muscles were isolated from mice, washed in 1x PBS for 5 minutes and stained for β-gal expression. Whole muscle tissues were fixed with 0.5% glutaraldehyde and immersed in 5-bromo-4-chloro-3-indole-β-D-galactopyranoside (X-gal) staining solution (5 mM K3Fe(CN)6, 5 mM K4Fe(CN)6, 1 mg/ml X-gal and 1 mM MgCl2) for 1– 2 hours at 37°C. Stained tissues were imaged using a Fujipix 5400 camera.
Cryosections
The tissues that were analyzed were tibialis anterior, quadriceps, gastrocnemius, forelimb muscle, abdominal muscle, intercostal muscle, diaphragm, heart, liver, lung, spleen, and kidney. Tissue samples were snap-frozen in 2-methylbutane cooled with dry ice. Half of the sample was used for preparing cryo-sections using a cryostat (HM 550, Richard-Allan Scientific) and the other half was used to extract protein and DNA. The cryosections taken on glass slides were stained for β-gal expression. Sections were fixed with 0.5% glutaraldehyde, stained in X-gal staining solution (5 mM K3Fe(CN)6, 5 mM K4Fe(CN)6, 1 mg/ml X-gal and 1 mM MgCl2) for 2– 3 hours at 37°C, mounted and viewed with a Zeiss Axiophot microscope.
Immunohistochemistry
Tibialis anterior muscle sections were pretreated with a blocking solution (Mouse-on Mouse [M.O.M.]; Vector, Burlingame, CA) in order to prevent nonspecific antibody binding. Sections were then incubated with polyclonal rabbit anti-β-galactosidase (5′ Prime 3′ Prime, Inc., USA) diluted at 1:2000 and monoclonal anti-myosin (skeletal, slow) (Sigma, USA) diluted at 1:2000 for 2 hrs. After a 20 min wash in 1X phosphate buffered saline (PBS), the sections were incubated with a Cy3 AffiniPure Donkey Anti-Rabbit IgG (Jackson ImmunoResearch Inc., USA) diluted 1:300 with M.O.M. and an Alexa Fluor® 488 goat anti-mouse IgG (Invitrogen, USA) diluted 1:100 with M.O.M. for 30 min at room temperature. Cell nuclei were stained with Hoechst. Muscle sections were visualized using fluorescence microscopy.
Ortho-nitro-phenyl galactopyranoside (ONPG) β-gal expression assay
To quantitate β-gal expression, the ONPG assay was performed on extracted protein from tissue samples.23 Briefly, snap frozen tissue samples were treated with TEES buffer (25 mM Tris-HCl pH 8.0, 2.5 mM ethylenediaminetetraacetic acid [EDTA] pH 8.0, 2.5 mM ethyleneglycoltetraacetic acid [EGTA] pH 7.4, 5% sodium dodecyl sulfate [SDS]) on ice. Samples were then centrifuged at 14,000 rpm for 30 min and protein extracts were collected and stored at −80°C until analysis. Subsequently, the protein extracts were used for ONPG assay and bicinchoninic acid (BCA) protein assay (Pierce, Rockford, IL). For the ONPG assay, serial dilutions of the protein extract were incubated at 37°C for 30 min in a 96-well plate with a buffer containing the enzyme substrate for the reaction. The optical density of the reaction was then read at wavelength 420 nm. The quantified samples were presented as units of β-gal activity/ng of protein.
Real time PCR assay
Total DNA was isolated from the muscle tissue by ethanol precipitation (Wizard genomic DNA purification kit, Promega, USA). Viral genomes were quantified in DNA from tissue samples using real-time PCR as previously described.23 Briefly, a 50 μl PCR volume contained 10 μl of DNA, 200 nM of each primer, 200 nM probe and 25 μl of TaqMan Universal Master Mix (PE Applied Biosystem, Foster City, CA, USA) containing 8% glycerol, 1X TaqMan buffer A, 5 mM MgCl2, 400μM dUTP, 200μM dATP, dCTP, dGTP (each), AmpliTaq Gold (0.025 U/μl) and AmpErase UNG (0.01 U/μl). The vector genomes were calculated by amplifying lacZ and normalized to endogenous mouse apolipoprotein B (Apo-B) (a single copy gene used as an internal control) used to calculate the amount of DNA (and thus the number of nuclei) in each sample. The primers and probes for the lacZ24 and Apo B25 genes have been previously described. The amount of DNA for each sample was calculated from the number of nuclei in each sample using the approximation that a murine diploid nucleus contains 6 pg of DNA. All real-time PCR assays were performed in MicroAmp optical 96-well reaction plates (PE Applied Biosystem, USA). Amplification conditions of 2 min at 50°C and 10 min at 95°C for the first cycle, followed by 40 cycles of 95°C for 15 s and 60°C for 1 min were used. The results were presented as copies of vector particles per 1000 nuclei.
Results
Tissue biodistribution of lacZ expression after intraperitoneal administration of AAV8lacZ in utero
To determine the biodistribution and gene transfer efficiency of systemically delivered AAV8 in utero, 6.4 × 1011 vector genomes of AAV8lacZ were injected IP per fetus on E-16. At 9 weeks of age, tissues were collected from injected pups, uninjected littermates and mothers carrying the treated pups. To assess the results of gene delivery, all tissues were analyzed for the expression of β-gal by X-gal staining. By whole tissue staining, diaphragm, intercostal muscles and abdominal muscles from the injected pups had the highest levels of β-gal expression (Figure 1). Moderate levels of expression were seen in hindlimb muscles, forelimb muscles and heart. Minimal to undetectable levels of expression were observed in liver, lung, spleen and kidney (data not shown).
Figure 1. β-galactosidase (β-gal) expression in skeletal and cardiac muscle tissues.
Tissues were collected 9 weeks after an intraperitoneal injection of 6.4 × 1011 vector genomes of AAV8lacZ into E-16 pups of pregnant CD1 mice and stained for β-gal expression using X-gal. X-gal staining was observed in hindlimb (a), forelimb (b), heart (c), abdominal muscles (d), diaphragm (e), and intercostal muscles (f).
Histological analysis of lacZ expression after intraperitoneal administration of AAV8lacZ in utero
Cryosections of diaphragm, quadriceps, gastrocnemius, tibialis anterior, forelimb muscle and heart collected from injected pups at 9 weeks of age were stained for β-gal expression. Diaphragm exhibited β-gal expression in nearly all muscle fibers (Figure 2b). A mosaic pattern of muscle fibers expressing β-gal was observed in forelimb, tibialis anterior, quadriceps and gastrocnemius muscles (Figure 2). A mosaic pattern of β-gal expressing cells was observed in cross-sections of heart with most transgene-expressing cells in proximity to the ventricles.
Figure 2. β-galactosidase (β-gal) expression in cryosections of muscle and heart.
Tissues were sectioned and stained for β-gal 9 weeks following an intraperitoneal injection of 6.4 × 1011 vector genomes of AAV8lacZ into E-16 pups of pregnant CD1 mice. β-gal expression was observed in cryo-sections of forelimb (a), diaphragm (b), heart (c), tibialis anterior (d), quadriceps (e), and gastrocnemius (f) muscles. Scale bar = 200 μm.
Quantification of β-gal protein expression in various tissues after intraperitoneal administration of AAV8lacZ in utero
In order to quantify the expression of protein we performed the ONPG assay on protein extracts from all tissues. Diaphragm and intercostal muscles exhibited high levels of expression of 7.73 ± 1.63 and 6.48 ± 0.94 units of β-gal per ng of protein respectively (Figure 3a). Quadriceps, tibialis anterior, gastrocnemius and forelimb muscle, and heart had moderate levels of expression of 2.2 ± 0.46, 1.40 ± 0.28, 1.91 ± 0.41, 2.47 ± 0.43, and 1.39 ± 0.29 units of β-gal per ng of protein respectively (Figure 3b). Liver, lung, kidney and spleen showed very low levels of expression of 0.51 ± 0.21, 0.84 ± 0.13, 0.22 ± 0.11, and 0.14 ± 0.08 units of β-gal per ng of protein respectively (Figure 3b). There were no detectable levels of β-gal expression in diaphragm, intercostal muscles, quadriceps, gastrocnemius, tibialis anterior, forelimb, heart, liver, lung, spleen and kidney tissues collected from the mothers that carried the treated mice in utero (data not shown).
Figure 3. Biodistribution of vector particles of AAV8lacZ and expression of β-galactosidase (β-gal).
Tissues were collected 9 weeks after an intraperitoneal injection of 6.4 × 1011 vector genomes of AAV8lacZ into E-16 pups of pregnant CD1 mice. Viral particles and β-gal expression were quantified by real-time PCR and ONPG assay, respectively, in diaphragm and intercostal muscles (a) and quadriceps, tibialis anterior, gastrocnemius, forelimb muscle, heart, liver, lung, kidney and spleen (b). Quantification of viral particles is expressed as mean viral particles per 1000 nuclei. Quantification of β-gal expression is shown as units of β-gal per ng of protein. Error bars represent standard error; number of mice analyzed (n) = 8. Viral particles per 1000 nuclei ■; Units of β-gal per ng protein .
Quantification of AAV vector genomes in various tissues after intraperitoneal administration of AAV8lacZ in utero
To determine viral vector gene transfer efficiency, we quantified the number of viral particles in individual tissues by real time PCR. The highest levels of viral vector particles were observed in diaphragm and intercostal muscles. Diaphragm and intercostal muscles had 294.68 ± 51.13 and 251.76 ± 83.28 viral particles per 1000 nuclei respectively (Figure 3a). Quadriceps, tibialis anterior, gastrocnemius, forelimb muscle, and heart had 37.20 ± 8.92, 15.52 ± 3.03, 23.24 ± 5.31, 62.72 ±19.40, and 13.80 ± 4.13 viral particles per 1000 nuclei respectively (Figure 3b). We also quantified the number of viral particles in non-muscle tissues. Liver, lung, kidney, and spleen had 3.79 ± 1.28, 3.76 ± 1.28, 3.76 ± 1.35, and 1.32 ± 0.49 viral particles per 1000 nuclei respectively (Figure 3b). There were no detectable levels of vector genomes in diaphragm, intercostal muscles, quadriceps, gastrocnemius, tibialis anterior, forelimb, heart, liver, lung, spleen and kidney tissues collected from the mothers that carried the treated mice in utero (data not shown).
Systemic administration of AAV8lacZ in utero demonstrates preferential transduction of fast-twitch muscle fibers
To better understand the distribution of muscle fibers transduced by AAV8lacZ in utero, we correlated transduction efficiency with muscle fiber type. Myofibers can be classified as fast-twitch or slow-twitch based on the isoform of myosin heavy chain (MyHC) expressed.26,27 Previous studies have shown that AAV2 preferentially transduced slow-twitch fibers and AAV6 transduced both fiber types in mice.28–30 Similarly another study showed AAV9 preferentially transduced fast-twitch fibers in mice;31 however AAV9 did not show any fiber type transduction preference in dogs.32 To determine if the transduction by AAV8lacZ in utero correlated with fiber type we performed double immunostaining with antibodies raised against β-gal and slow-twitch MyHC. Interestingly, we observed that the majority of β-gal-expressing muscle fibers did not express slow-twitch MyHC. However, rare slow-twitch fibers were transduced (Figure 4).
Figure 4. Expression of β-galactosidase (β-gal) in slow- and fast-twitch muscle fibers.
Section of tibialis anterior muscle from a 9-week-old CD1 mouse treated in utero at E-16 with intraperitoneal administration of 6.4 × 1011 vector genomes of AAV8lacZ was immunostained for β-gal (red, β-gal) and slow-twitch myosin heavy chain (green, Slow). The merge image shows that most transduced fibers were fast-twitch fibers (examples labeled with #). Rare slow-twitch fibers were also transduced by AAV8lacZ (examples labeled with *). Nuclei are stained with Hoechst. Scale bar = 200 μm.
Discussion
In this study, we systemically delivered AAV8 vector to E-16 mice in utero and analyzed the distribution of transgene expression and vector genomes in treated mice at 9 weeks of age. While considerable AAV8 gene therapy studies have been done in adult and neonatal mice, the biodistribution of AAV8 vector after in utero gene delivery has not been reported. We here provide the first report that widespread gene expression in various muscles, including forelimb, diaphragm, intercostal, heart, tibialis anterior, quadriceps, and gastrocnemius muscles after AAV8 lacZ in utero injection. In addition to demonstrating lacZ gene expression by X-gal staining, we quantified gene expression by ONPG assay, and quantified the number of gene copies by real time PCR. We observed a good correlation between the level of transgene protein expression and vector genome copy numbers in individual tissues. In particular, we observed highest levels of expression in diaphragm and intercostal muscles followed by other skeletal muscles. Interestingly, we noted minimal vector transduction in non-muscle tissues such as the liver, lung, spleen and kidney. Importantly, we observed no detectable levels of β-gal expression (by ONPG) or vector genomes (by Taqman real time PCR) in tissues collected from the mothers that carried the experimental mice in utero. Maternal tissues studied included diaphragm, intercostal muscles, quadriceps, gastrocnemius, tibialis anterior, forelimb, heart, liver, lung, spleen and kidney
Previous studies have shown the potential of AAV8 vectors to deliver transgenes to muscle tissue of postnatal mice.2–9 With systemic administration, AAV8 vector exhibited better transduction in neonates than adults in mice suggesting systemic barriers to gene delivery that develop with maturation of adult tissues.7 This suggested that in utero gene delivery of AAV8 would offer even more efficient gene delivery with potential benefit for the treatment of genetic diseases in particular. Consistent with previous postnatal AAV8 vector studies we observed high gene transduction in skeletal muscles with in utero AAV8 vector gene transfer. In this respect, we noted a widespread and mosaic pattern of gene expression throughout various muscle groups both in the upper and lower limbs. Importantly, a high level of gene transduction was observed in diaphragm and intercostal muscles when gene delivery of AAV8 vector was accomplished in utero.
Some parallels can be drawn between gene delivery in utero and to neonatal mice by comparing our results with a prior AAV8 systemic delivery study in neonatal mice published by Wang et al.7 In this study 1-day-old mouse pups treated with IP administration of AAV8 showed high transduction efficiency in various muscle tissues for up to 2 months. The dose and period of analysis of the neonatal study were comparable to our in utero study. Similar to the neonatal study, we observed expression in various muscle tissues, heart and diaphragm. Furthermore, similar to the neonatal study, our study also showed minimal to undetectable expression in non-muscle tissues such as liver, lung, kidney and spleen.
Previous studies of viral vectors injected in utero have exhibited promising, but varying abilities to deliver transgenes to different tissues.19,20,33,34 Prior in utero viral vector-mediated gene delivery studies have been done with AAV1 and AAV2,19,20,35,36 AAV5,36 adenoviral vectors,33,36,37 and lentiviral vectors.38 While some of the previous in utero studies using viral vectors such as AAV1,19 AAV219 and AAV536 demonstrated minimal skeletal muscle expression, our study using AAV8 in utero systemic gene delivery demonstrated significant levels of widespread gene delivery to skeletal muscles including forelimbs and hindlimbs. Furthermore, the high level of gene transduction in respiratory muscles was accomplished with a relatively low dose of AAV8 vector.
Different patterns of association of gene transduction to muscle fiber type have been observed for different AAV serotypes. While studies have shown AAV6 transduces both fast- and slow-twitch fibers in young adult mice (5–6 week old C57BL/6) in extensor digitorum longus (EDL) and soleus muscles29 but with a trend towards preferential transduction of fast-twitch fibers treated in 3-day-old mdx hind limb muscle,28 it has also been shown that AAV2 preferentially transduces slow-twitch fibers in hindlimb muscle of both newborn and adult mice.30 Moreover, studies with AAV9 demonstrated that both fiber types were transduced in gastocnemius muscle of newborn dogs and that AAV9 transduction was independent of laminin receptor (LamR) expression,32 a known receptor of AAV9.39 In contrast, studies in soleus and TA muscles of newborn and young adult (7 week old) C57BL/10 mice demonstrated that AAV9 preferentially transduced fast-twitch fibers.31 Hence to further understand the transduction profile of AAV8 in utero we studied the fiber type transduction in TA muscle. We observed preferential transduction of fast-twitch fibers. However, we also noted transduction of rare slow-twitch fibers. Further studies will be required to understand the reasons for fiber type preference in transduction that appears to depend upon vector serotype and host factors such as species and age.
Studies have shown that many viral vectors injected systemically into adult mice preferentially transduce the liver.2,7 However, our in utero gene delivery data, consistent with other in utero studies,19,33,34 demonstrated minimal transduction of viral vectors into liver. It is interesting to note that even when delivered by the intrahepatic route, in utero injection of AAV2 provided minimal liver transduction (<1%) at days 21 and 70 post treatment in mice.20 Similarly intramniotoic injection of AAV2 to transduce rabbit fetuses demonstrated no transduction into liver tissue.34 These and other studies19,33 demonstrated very low transduction of the liver by in utero gene transfer. This possibly is due to high cell turn over and unique properties in the fetal liver compared to the adult liver.40–42 From the standpoint of safety this is significant since liver transduction often leads to toxicity and has been an important limitation of clinical viral vector gene transfer studies.43 Furthermore, liver transduction is generally not desired as a component of muscle gene transfer strategies.
Studies have shown that IP injections of vectors such as AAV1 and AAV2 have varying abilities to transduce the murine heart in utero.19,44 Lipshutz et al. showed that while the gene expression of AAV2 in the peritoneum of CD1 mice continued to persist up to 18 months post in utero transuterine IP injection, the expression in heart declined.44 Similarly, it has been shown that while the diaphragm showed high transgene expression for up to 4 weeks after intraperitoneal in utero treatment of AAV1, the heart demonstrated minimal expression in C57BL/6 mice.19 Hence in order to understand the transduction ability of AAV8 vector in heart after in utero gene delivery, we performed X-gal staining and quantitative assessment of β-gal expression. We observed that although the transgene expression levels in heart did not reach as high as most limb muscles we did observe β-gal-expressing cells in close proximity to the ventricles with levels approaching that of the limb muscle tissues.
Given that many muscular dystrophy patients require ventilatory assistance and many die due to respiratory failure, the high amount of AAV8 vector transduction observed in respiratory muscles offers the potential to provide functional benefit to these patients. In addition to disorders due to deficiencies of muscle membrane proteins such as Duchenne muscular dystrophy, AAV8 also has potential as a gene delivery vector in other genetic muscle diseases where patients die due to respiratory failure such as Pompe disease.45 Patients suffering from infantile-onset Pompe disease start showing symptoms at a median age of 1.6 months and die at a median age of 6–8 months.45 Thus the significance of effective vector transduction into respiratory muscles prenatally could potentially help improve the clinical condition of these patients.
In this report we demonstrate the potential of systemic delivery of AAV8 vector in utero to achieve widespread muscle transduction with especially high levels in respiratory muscles. The findings suggest the therapeutic potential for delivering genes systemically to muscle cells of disease models by in utero gene delivery of AAV8 vectors.
Acknowledgments
This work was supported by Grant R01 AR050565 (PRC) and R01 AR45967 (XX) from the NIH. This material is also the result of work supported by VA resources (VA Healthcare System, Pittsburgh, PA, USA).
Footnotes
Work was done in Pittsburgh, PA, USA.
References
- 1.Xiao X, Li J, Samulski RJ. Efficient long-term gene transfer into muscle tissue of immunocompetent mice by adeno-associated virus vector. J Virol. 1996;70:8098–8108. doi: 10.1128/jvi.70.11.8098-8108.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Zincarelli C, Soltys S, Rengo G, Rabinowitz JE. Analysis of AAV serotypes 1–9 mediated gene expression and tropism in mice after systemic injection. Mol Ther. 2008;16:1073–1080. doi: 10.1038/mt.2008.76. [DOI] [PubMed] [Google Scholar]
- 3.Ziegler RJ, Bercury SD, Fidler J, Zhao MA, Foley J, Taksir TV, et al. Ability of Adeno-Associated Virus Serotype 8-Mediated Hepatic Expression of Acid alpha-Glucosidase to Correct the Biochemical and Motor Function Deficits of Presymptomatic and Symptomatic Pompe Mice. Hum Gene Ther. 2008 doi: 10.1089/hum.2008.010. [DOI] [PubMed] [Google Scholar]
- 4.Qiao C, Li J, Jiang J, Zhu X, Wang B, Li J, et al. Myostatin propeptide gene delivery by adeno-associated virus serotype 8 vectors enhances muscle growth and ameliorates dystrophic phenotypes in mdx mice. Hum Gene Ther. 2008;19:241–254. doi: 10.1089/hum.2007.159. [DOI] [PubMed] [Google Scholar]
- 5.Rodino-Klapac LR, Janssen PM, Montgomery CL, Coley BD, Chicoine LG, Clark KR, et al. A translational approach for limb vascular delivery of the micro-dystrophin gene without high volume or high pressure for treatment of Duchenne muscular dystrophy. J Transl Med. 2007;5:45. doi: 10.1186/1479-5876-5-45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Inagaki K, Fuess S, Storm TA, Gibson GA, McTiernan CF, Kay MA, et al. Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Mol Ther. 2006;14:45–53. doi: 10.1016/j.ymthe.2006.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Wang Z, Zhu T, Qiao C, Zhou L, Wang B, Zhang J, et al. Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat Biotechnol. 2005;23:321–328. doi: 10.1038/nbt1073. [DOI] [PubMed] [Google Scholar]
- 8.Louboutin JP, Wang L, Wilson JM. Gene transfer into skeletal muscle using novel AAV serotypes. J Gene Med. 2005;7:442–451. doi: 10.1002/jgm.686. [DOI] [PubMed] [Google Scholar]
- 9.Sun B, Zhang H, Franco LM, Young SP, Schneider A, Bird A, et al. Efficacy of an adeno-associated virus 8-pseudotyped vector in glycogen storage disease type II. Mol Ther. 2005;11:57–65. doi: 10.1016/j.ymthe.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 10.Vandenberghe LH, Wang L, Somanathan S, Zhi Y, Figueredo J, Calcedo R, et al. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med. 2006;12:967–971. doi: 10.1038/nm1445. [DOI] [PubMed] [Google Scholar]
- 11.Thomas CE, Storm TA, Huang Z, Kay MA. Rapid uncoating of vector genomes is the key to efficient liver transduction with pseudotyped adeno-associated virus vectors. J Virol. 2004;78:3110–3122. doi: 10.1128/JVI.78.6.3110-3122.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Davidoff AM, Gray JT, Ng CY, Zhang Y, Zhou J, Spence Y, et al. Comparison of the ability of adeno-associated viral vectors pseudotyped with serotype 2, 5, and 8 capsid proteins to mediate efficient transduction of the liver in murine and nonhuman primate models. Mol Ther. 2005;11:875–888. doi: 10.1016/j.ymthe.2004.12.022. [DOI] [PubMed] [Google Scholar]
- 13.Nathwani AC, Gray JT, Ng CY, Zhou J, Spence Y, Waddington SN, et al. Self-complementary adeno-associated virus vectors containing a novel liver-specific human factor IX expression cassette enable highly efficient transduction of murine and nonhuman primate liver. Blood. 2006;107:2653–2661. doi: 10.1182/blood-2005-10-4035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nathwani AC, Gray JT, McIntosh J, Ng CY, Zhou J, Spence Y, et al. Safe and efficient transduction of the liver after peripheral vein infusion of self-complementary AAV vector results in stable therapeutic expression of human FIX in nonhuman primates. Blood. 2007;109:1414–1421. doi: 10.1182/blood-2006-03-010181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Cheng H, Wolfe SH, Valencia V, Qian K, Shen L, Phillips MI, et al. Efficient and persistent transduction of exocrine and endocrine pancreas by adeno-associated virus type 8. J Biomed Sci. 2007;14:585–594. doi: 10.1007/s11373-007-9159-1. [DOI] [PubMed] [Google Scholar]
- 16.Foust KD, Poirier A, Pacak CA, Mandel RJ, Flotte TR. Neonatal intraperitoneal or intravenous injections of recombinant adeno-associated virus type 8 transduce dorsal root ganglia and lower motor neurons. Hum Gene Ther. 2008;19:61–70. doi: 10.1089/hum.2007.093. [DOI] [PubMed] [Google Scholar]
- 17.Nakai H, Fuess S, Storm TA, Muramatsu S, Nara Y, Kay MA. Unrestricted hepatocyte transduction with adeno-associated virus serotype 8 vectors in mice. J Virol. 2005;79:214–224. doi: 10.1128/JVI.79.1.214-224.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Graham T, McIntosh J, Work LM, Nathwani A, Baker AH. Performance of AAV8 vectors expressing human factor IX from a hepatic-selective promoter following intravenous injection into rats. Genet Vaccines Ther. 2008;6:9. doi: 10.1186/1479-0556-6-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bilbao R, Reay DP, Li J, Xiao X, Clemens PR. Patterns of gene expression from in utero delivery of adenoviral-associated vector serotype 1. Hum Gene Ther. 2005;16:678–684. doi: 10.1089/hum.2005.16.678. [DOI] [PubMed] [Google Scholar]
- 20.Mitchell M, Jerebtsova M, Batshaw ML, Newman K, Ye X. Long-term gene transfer to mouse fetuses with recombinant adenovirus and adeno-associated virus (AAV) vectors. Gene Therapy. 2000;7:1986–1992. doi: 10.1038/sj.gt.3301332. [DOI] [PubMed] [Google Scholar]
- 21.Xiao X, Li J, Samulski RJ. Production of high-titer recombinant adeno-associated virus vectors in the absence of helper adenovirus. J Virol. 1998;72:2224–2232. doi: 10.1128/jvi.72.3.2224-2232.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li J, Samulski RJ, Xiao X. Role for highly regulated rep gene expression in adeno-associated virus vector production. J Virol. 1997;71:5236–5243. doi: 10.1128/jvi.71.7.5236-5243.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bilbao R, Reay DP, Hughes T, Biermann V, Volpers C, Goldberg L, et al. Fetal muscle gene transfer is not enhanced by an RGD capsid modification to high-capacity adenoviral vectors. Gene Ther. 2003;10:1821–1829. doi: 10.1038/sj.gt.3302084. [DOI] [PubMed] [Google Scholar]
- 24.Senoo M, Matsubara Y, Fujii K, Nagasaki Y, Hiratsuka M, Kure S, et al. Adenovirus-mediated in utero gene transfer in mice and guinea pigs: tissue distribution of recombinant adenovirus determined by quantitative TaqMan-polymerase chain reaction assay. Mol Genet Metab. 2000;69:269–276. doi: 10.1006/mgme.2000.2984. [DOI] [PubMed] [Google Scholar]
- 25.Pan D, Gunther R, Duan W, Wendell S, Kaemmerer W, Kafri T, et al. Biodistribution and toxicity studies of VSVG-pseudotyped lentiviral vector after intravenous administration in mice with the observation of in vivo transduction of bone marrow. Mol Ther. 2002;6:19–29. doi: 10.1006/mthe.2002.0630. [DOI] [PubMed] [Google Scholar]
- 26.Reiser PJ, Greaser ML, Moss RL. Myosin heavy chain composition of single cells from avian slow skeletal muscle is strongly correlated with velocity of shortening during development. Dev Biol. 1988;129:400–407. doi: 10.1016/0012-1606(88)90387-9. [DOI] [PubMed] [Google Scholar]
- 27.Barany M. ATPase activity of myosin correlated with speed of muscle shortening. J Gen Physiol. 1967;50(Suppl):197–218. doi: 10.1085/jgp.50.6.197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lai Y, Yue Y, Liu M, Ghosh A, Engelhardt JF, Chamberlain JS, et al. Efficient in vivo gene expression by trans-splicing adeno-associated viral vectors. Nat Biotechnol. 2005;23:1435–1439. doi: 10.1038/nbt1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Blankinship MJ, Gregorevic P, Allen JM, Harper SQ, Harper H, Halbert CL, et al. Efficient transduction of skeletal muscle using vectors based on adeno-associated virus serotype 6. Mol Ther. 2004;10:671–678. doi: 10.1016/j.ymthe.2004.07.016. [DOI] [PubMed] [Google Scholar]
- 30.Pruchnic R, Cao B, Peterson ZQ, Xiao X, Li J, Samulski RJ, et al. The use of adeno-associated virus to circumvent the maturation-dependent viral transduction of muscle fibers. Hum Gene Ther. 2000;11:521–536. doi: 10.1089/10430340050015716. [DOI] [PubMed] [Google Scholar]
- 31.Bostick B, Ghosh A, Yue Y, Long C, Duan D. Systemic AAV-9 transduction in mice is influenced by animal age but not by the route of administration. Gene Ther. 2007;14:1605–1609. doi: 10.1038/sj.gt.3303029. [DOI] [PubMed] [Google Scholar]
- 32.Yue Y, Ghosh A, Long C, Bostick B, Smith BF, Kornegay JN, et al. A single intravenous injection of adeno-associated virus serotype-9 leads to whole body skeletal muscle transduction in dogs. Mol Ther. 2008;16:1944–1952. doi: 10.1038/mt.2008.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bilbao R, Reay DP, Wu E, Zheng H, Biermann V, Kochanek S, et al. Comparison of high-capacity and first-generation adenoviral vector gene delivery to murine muscle in utero. Gene Ther. 2005;12:39–47. doi: 10.1038/sj.gt.3302392. [DOI] [PubMed] [Google Scholar]
- 34.Boyle MP, Enke RA, Adams RJ, Guggino WB, Zeitlin PL. In utero AAV-mediated gene transfer to rabbit pulmonary epithelium. Mol Ther. 2001;4:115–121. doi: 10.1006/mthe.2001.0428. [DOI] [PubMed] [Google Scholar]
- 35.Sabatino DE, Mackenzie TC, Peranteau W, Edmonson S, Campagnoli C, Liu YL, et al. Persistent expression of hF.IX After tolerance induction by in utero or neonatal administration of AAV-1-F. IX in hemophilia B mice. Mol Ther. 2007;15:1677–1685. doi: 10.1038/sj.mt.6300219. [DOI] [PubMed] [Google Scholar]
- 36.Bouchard S, MacKenzie TC, Radu AP, Hayashi S, Peranteau WH, Chirmule N, et al. Long-term transgene expression in cardiac and skeletal muscle following fetal administration of adenoviral or adeno-associated viral vectors in mice. J Gene Med. 2003;5:941–950. doi: 10.1002/jgm.421. [DOI] [PubMed] [Google Scholar]
- 37.Reay DP, Bilbao R, Koppanati BM, Cai L, O’Day TL, Jiang Z, et al. Full-length dystrophin gene transfer to the mdx mouse in utero. Gene Ther. 2008;15:531–536. doi: 10.1038/gt.2008.8. [DOI] [PubMed] [Google Scholar]
- 38.MacKenzie TC, Kobinger GP, Louboutin JP, Radu A, Javazon EH, Sena-Esteves M, et al. Transduction of satellite cells after prenatal intramuscular administration of lentiviral vectors. J Gene Med. 2005;7:50–58. doi: 10.1002/jgm.649. [DOI] [PubMed] [Google Scholar]
- 39.Akache B, Grimm D, Pandey K, Yant SR, Xu H, Kay MA. The 37/67-kilodalton laminin receptor is a receptor for adeno-associated virus serotypes 8, 2, 3, and 9. J Virol. 2006;80:9831–9836. doi: 10.1128/JVI.00878-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Taylor PA, McElmurry RT, Lees CJ, Harrison DE, Blazar BR. Allogenic fetal liver cells have a distinct competitive engraftment advantage over adult bone marrow cells when infused into fetal as compared with adult severe combined immunodeficient recipients. Blood. 2002;99:1870–1872. doi: 10.1182/blood.v99.5.1870. [DOI] [PubMed] [Google Scholar]
- 41.O’Donoghue K, Fisk NM. Fetal stem cells. Best Pract Res Clin Obstet Gynaecol. 2004;18:853–875. doi: 10.1016/j.bpobgyn.2004.06.010. [DOI] [PubMed] [Google Scholar]
- 42.Lansdorp PM, Dragowska W, Mayani H. Ontogeny-related changes in proliferative potential of human hematopoietic cells. J Exp Med. 1993;178:787–791. doi: 10.1084/jem.178.3.787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Marshall E. Gene therapy death prompts review of adenovirus vector. Science. 1999;286:2244–2245. doi: 10.1126/science.286.5448.2244. [DOI] [PubMed] [Google Scholar]
- 44.Lipshutz GS, Gruber CA, Cao Y, Hardy J, Contag CH, Gaensler KM. In utero delivery of adeno-associated viral vectors: intraperitoneal gene transfer produces long-term expression. Mol Ther. 2001;3:284–292. doi: 10.1006/mthe.2001.0267. [DOI] [PubMed] [Google Scholar]
- 45.Winkel LP, Hagemans ML, van Doorn PA, Loonen MC, Hop WJ, Reuser AJ, et al. The natural course of non-classic Pompe’s disease; a review of 225 published cases. J Neurol. 2005;252:875–884. doi: 10.1007/s00415-005-0922-9. [DOI] [PubMed] [Google Scholar]