当前位置: 首页 > 期刊 > 《循环学杂志》 > 2005年第10期 > 正文
编号:11304616
Sustained Whole-Body Functional Rescue in Congestive Heart Failure and Muscular Dystrophy Hamsters by Systemic Gene Transfer
http://www.100md.com 《循环学杂志》
     the Molecular Therapy Laboratory, Department of Orthopedic Surgery (T.Z., L.Z., Z.W., C.Q., C.C., J.L., X.X.), Department of Molecular Genetics and Biochemistry (X.X.), and Cardiovascular Institute (S.M., C.F.M.), University of Pittsburgh School of Medicine, Pittsburgh, Pa, and the Department of Cardiology (C.C., D.W.W.), Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.

    Abstract

    Background— The success of muscular dystrophy gene therapy requires widespread and stable gene delivery with minimal invasiveness. Here, we investigated the therapeutic effect of systemic delivery of adeno-associated virus (AAV) vectors carrying human -sarcoglycan (-SG) gene in TO-2 hamsters, a congestive heart failure and muscular dystrophy model with a -SG gene mutation.

    Methods and Results— A single injection of double-stranded AAV serotype 8 vector carrying human -SG gene without the need of any physical or pharmaceutical interventions achieved nearly complete gene transfer and tissue-specific expression in the heart and skeletal muscles of the diseased hamsters. Broad and sustained (>12 months) restoration of the missing -SG gene in the TO-2 hamsters corrected muscle cell membrane leakiness throughout the body and normalized serum creatine kinase levels (a 50- to 100-fold drop). Histological examination revealed minimal or the absence of central nucleation, fibrosis, and calcification in the skeletal muscle and heart. Whole-body functional analysis such as treadmill running showed dramatic improvement, similar to the wild-type F1B hamsters. Furthermore, cardiac functional studies with echocardiography revealed significantly increased percent fractional shortening and decreased left ventricular end-diastolic and end-systolic dimensions in the treated TO-2 hamsters. The survival time of the animals was also dramatically extended.

    Conclusions— Systemic gene transfer of -SG by the AAV serotype 8 vector could effectively ameliorate cardiac and skeletal muscle pathology, profoundly improve cardiac and whole-body functions, and significantly prolong the lifespan of the treated TO-2 hamsters.

    Key Words: viruses cardiomyopathy gene therapy

    Introduction

    Muscular dystrophies are common and debilitating genetic diseases that afflict a significant population of children and adults worldwide. No effective treatment is currently available. Besides the loss of limb functions, progressive muscle deterioration in the patients eventually leads to respiratory and heart failure. These diseases affect the striated muscles throughout the body, both skeletal and cardiac. Efficient whole-body delivery of the therapeutic genes or stem cells to correct the genetic defects has been sought.1–5 Gene vectors derived from the nonpathogenic and replication-defective adeno-associated viruses (AAV) are among the most promising systems for muscle and heart gene delivery.6–11 Local intramuscular injection has demonstrated the effectiveness of AAV vector-mediated gene transfer.2,6,12–16 The blood circulation system could serve as a natural conduit for systemic gene delivery, but the capillary vasculature in the muscle tissues imposes a major physical and biological barrier limiting gene vector dissemination. As a result, both physical17,18 and pharmaceutical methods6,10,19,20 have been developed to overcome the barrier. These methods, however, are either limited to limb muscles or associated with severe invasiveness and/or pharmaceutical side effects.21 The discovery of novel serotypes of AAV22–27 has prompted us to explore the utility of these new serotypes for systemic gene delivery to muscle without invasive interventions. Findings from a recent comparison of an array of vectors derived from the novel AAV serotypes3 suggest that AAV serotype 8 (AAV8) is the most efficient in crossing the capillary blood vessels to achieve systemic gene delivery in heart and muscles. The cardiomyopathy Syrian hamster Bio14.628 and its derivative strains, including TO-2, are the first available limb girdle muscular dystrophy (LGMD) animal model. These hamsters are broadly used in studies for congestive heart failure, muscular dystrophy, small-molecule drug therapy, and gene therapy.6,10,14,29–32 With a large deletion in the -sarcoglycan (SG) gene,33,34 the hamsters manifest pathologies that closely mimic the severe and early-onset forms of LGMD,35 which are usually caused by mutations in the genes encoding SGs, including SG- (LGMD 2D), SG- (LGMD 2E), SG- (LGMD 2C), and SG- (LGMD 2F). These small transmembrane proteins are found mainly on both skeletal and cardiac muscle cells. The SGs associate in equal stoichiometry to form a heterotetramer, named SG complex.36 Primary deficiency of any single SG protein will result in partial or complete disappearance of the SG complex on the striated muscle cell membrane, leading to the disease phenotypes. Besides the skeletal muscle myopathy, the -SG–deficient hamster TO-2 substrain in particular suffers from severe cardiac dilation and congestive heart failure, which is a primary cause of the much-shortened lifespan of those animals. Because of the resemblance to human patients, the TO-2 hamsters provide an excellent animal model for us to test the systemic gene therapy strategy for both skeletal and cardiac deficiencies.

    Clinical Perspective p 2659

    In this study, we have investigated the gene transfer efficiency and global therapeutic efficacy after systemic delivery of human -SG gene to the TO-2 hamsters by the AAV8 vectors. We found that extensive and sustained gene transfer in the cardiac and skeletal muscles throughout the body could be achieved by a single injection of AAV8 in neonatal hamsters via the intraperitoneal route or in adult hamsters via the intravenous route. Dramatic improvement in biochemical and histological parameters of the heart and muscle was observed as a result of systemic gene delivery of the therapeutic gene. In addition, cardiac and whole-body functions were also profoundly improved, accompanied by prolongation of lifespan of the treated TO-2 hamsters.

    Methods

    AAV Vector Construction and Production

    Recombinant double-stranded AAV vectors containing human SG cDNA driven by the cytomegalovirus (CMV) promoter or by the synthetic muscle-specific promoter37 (SP) C5–27 were constructed by the standard cloning protocols. The vector DNA was packaged into AAV2 or AAV8 viral particles with the commonly used triple-plasmid transfection method38 and purified twice by the standard CsCl density ultracentrifugation technique. The titers of the purified viral stocks were determined by DNA dot blot method in the range of 2 to 5x1012 vector genomes per milliliter.

    Animals and Vector Administration

    All experiments involving wild-type F1B and dystrophic TO-2 hamsters (Bio Breeders, Fitchburg, Mass) were approved by the University of Pittsburgh Animal Care and Use Committee. For vector administration, the neonatal TO-2 hamsters (age, 10 days) were injected intraperitoneally with AAV2 or AAV8 CMV-SG vectors (1x1012 vector genomes), whereas the adult TO-2 hamsters (age, 1.5 months) were injected intravenously with the AAV8 SP-SG vector (1x1012 vector genomes).

    Histology and Serum Creatine Kinase Enzyme Assay

    Muscle or other tissues were collected at the indicated time points after vector injection. For detection of SGs, tissue samples were processed by cryo-thin-sections at 6- to 8-μm thickness. Detection of the -, -, - and -SG by immunofluorescent staining or by Western blotting was also described previously.14 To detect fibrosis and calcification, Masson’s trichrome staining and Von Kossa staining, respectively, were performed according to histology handbooks. For in vivo tests of muscle cell membrane integrity, Evans blue dye (10 mg/mL PBS) was injected into the jugular vein of hamsters at a dose of 0.1 mg/g body weight. The hamsters were euthanized 3 hours later, and muscles were collected and cryosectioned. Evans blue dye–positive myofibers, which show strong red fluorescence, were observed under the fluorescent microscope. Serum creatine kinase (CK) enzyme activities were assayed with a kinetic NADH formation kit (Diagnostic Chemicals Limited) according to the manufacturer’s instruction.

    Treadmill

    For the whole-body functional test, adult hamsters were placed on a motorized treadmill (Harvard Apparatus, model 7054) and run at a 15° downward angle at a rate of 15 m/min. The test was stopped when the animal spent >5 seconds sitting on the shock grid without attempting to reenter the treadmill.

    Echocardiography Analysis

    Cardiac functions were analyzed by transthoracic echocardiography as described39 with a 13-MHz probe and a SEQUOIA 512 echocardiography machine (Acuson). The F1B hamsters were anesthetized with 60 mg/kg and TO-2 hamsters with 50 mg/kg sodium pentobarbital by intraperitoneal injection for consistent heart rates. Two-dimensional targeted M-mode imaging was obtained from the short axis immediately below the level of the mitral valve. M-mode tracings were recorded and measurements (left ventricular end-systolic dimension, left ventricular end-diastolic dimension, heart rate, left ventricle posterior wall thickness, and percentage fractional shortening) were made as previously reported,39 with the operator blinded to hamster genotype and treatment.

    Statistical Analysis

    Statistic analysis was performed with Fisher’s t test. Differences were considered significant at P<0.01.

    Results

    Systemic and Long-Term SG Gene Transfer in Heart and Muscle by AAV8 in Neonatal Hamsters

    First, we investigated side by side the gene transfer efficiencies of the most commonly used AAV2 and the newly discovered AAV8 carrying a therapeutic gene, the human -SG, in the hamsters of TO-2 strain, a naturally occurring congestive heart failure and LGMD 2F animal model. Our results showed that a single injection of the AAV8-CMV-SG vector (1x1012 vector genomes) by the intraperitoneal route in the 10-day-old neonatal TO-2 hamsters achieved nearly complete gene transfer in the heart and all the skeletal muscles we examined in hamsters at 3 months of age (2.5 months after gene transfer). In contrast, the AAV2 vector achieved only very limited gene transfer in the heart and various muscles (Figure 1a), except in the diaphragm and abdominal muscles, which were proximal to the peritoneal cavity and therefore readily infected. In addition, the AAV8-mediated systemic human -SG gene transfer and expression were both widespread and persistent from 3 months (Figure 1a) to 6 and 12 months (Figure 1b) during the studies. Careful examination of cross sections of the entire hind-leg tibialis anterior (TA) muscle at 6 and 12 months after gene transfer revealed -SG expression in nearly 100% of the myofibers (Figure 1b). Overexpression of -SG was observed in a fraction of the heart and diaphragm muscle cells (Figure 1a), in which both the plasma membrane and the cell bodies were stained positive by an antibody specific to the human -SG. It was also noticeable that overexpression of human -SG in hamster heart persisted for at least 6 months, which contributed to the recruitment of the missing components of SG complex such as -SG onto the cardiomyocyte membrane (Figure 1b). The cardiac structure and morphology were normal and free of cell infiltration. Similar phenomena were also seen after local delivery in the heart and muscle.9,14 Throughout the duration of the experiments, no immune rejection and overt toxicity were observed by monitoring the general health, physical activities, and tissue histology of the treated hamsters. These results indicate that AAV8 is capable of systemic and long-term gene delivery in neonatal hamsters with high efficiency.

    Systemic and Long-Term SG Gene Transfer and Muscle-Specific Expression by AAV8 in Adult Hamsters

    We next investigated whether comparable efficiency of -SG gene transfer could be achieved in the adult TO-2 hamsters. Here, we chose to use a muscle-specific, synthetic promoter SP37 instead of the CMV promoter to drive the -SG gene, because AAV8 is also capable of long-term gene transfer to a number of non-muscle tissues, especially the adult liver.3,22,40 The AAV8 -SG vector driven by the synthetic promoter SP was injected intravenously (1x1012 vector genomes per animal) via the jugular vein in the 1.5-month-old adult TO-2 hamsters, which already manifested pathological signs of muscular dystrophy. Similar to the neonatal hamsters treated with AAV8-CMV-SG, the adult-treated hamsters also exhibited nearly complete and long-term -SG expression in the heart (Figure 2a, top) and all of the skeletal muscles examined (see below), including TA muscle (Figure 2a, bottom).

    As expected, -SG expression was muscle specific and could not be detected in nonmuscle tissues by either immunofluorescent staining or Western blotting (Figure 2b and 2c), despite the fact that the AAV -SG vector DNA was present in the liver at a very high copy number and variably in other nonmuscle tissues (Figure 2d). It is noteworthy that unlike the shortened form of muscle-specific CK promoters,19 the SP promoter was found highly active in both the heart and diaphragm. These results demonstrated that efficient and stable systemic -SG gene delivery in the heart and muscle of adult TO-2 hamsters could also be achieved and that striated muscle-specific gene expression could be obtained by use of the synthetic muscle promoter.

    Restoration of Muscle Cell Membrane Integrity and Amelioration of Muscle Pathology by Systemic Gene Transfer

    We next investigated whether systemic -SG in the muscle could render therapeutic effects at the cellular and histological levels. Immunofluorescent staining of -, -, -, and -SGs on the cross-thin-sections of the heart and muscle samples revealed that expression of the missing -SG gene restored the missing SG complex onto the plasma membrane of TO-2 hamsters, which were treated as either neonates (Figure 3a) or adults (see below). The recovery of the SG complex on the striated muscle cell membrane has corrected the underlying biochemical deficiency and consequently restored the integrity of the dystrophic muscle membrane, which otherwise would be very leaky. In vivo examination with Evans blue, a commonly used small-molecule dye for cell membrane integrity test, showed no leakage in the myofibers of AAV8-treated TO-2 hamsters but substantial leakage in those of untreated TO-2 hamsters (Figure 3b).

    Additional evidence of systemic restoration of the muscle cell membrane integrity was the dramatic diminution of a hallmark sign of the muscular dystrophy, thus the elevated serum levels of CK, which came from the leaky dystrophic muscle cells. When examined at 2, 5, and 8 months after AAV8 vector injection, the serum CK activities of the TO-2 hamsters treated at the neonatal age were found to be completely normalized, indistinguishable from the wild-type F1B hamsters. In contrast, the serum CK activities in the untreated, age-matched TO-2 hamsters were up to 100-fold higher (Figure 3c, left). More compellingly, the serum CK activities in the TO-2 hamsters, which were treated at the adult age, also declined >100-fold to normal levels and remained normal when examined at 1 and 5 months after vector injection (Figure 3c, right). Complete normalization of the serum CK activities further substantiated the remarkable gene transfer efficiency revealed earlier by immunofluorescent staining of -SG in the muscle (Figure 1). Thus, the histological and biochemical assays strongly suggest that AAV8 has efficiently and stably transferred the therapeutic -SG gene into the vast majority, if not all, of the striated muscle cells in the neonatal and adult hamsters.

    Systemic gene delivery in both neonatal and adult TO-2 hamsters was also found to systemically ameliorate muscle pathology such as muscle degeneration and fibrosis. In the hamsters treated at the neonatal age, muscle degeneration was completely prevented as a result of gene transfer in all examined muscles (Figure 4a [top] and 4c). The percentages of myofibers containing centrally localized nuclei (CN), a classic sign of muscle degeneration and regeneration, was very low (< 1%) in hamsters examined at 3.5 and 9 months of age, identical to those of age-matched wild-type F1B hamsters (Figure 4b). In contrast, the untreated age-matched TO-2 hamsters exhibited CN rates >70% (Figure 4b). Furthermore, in the TO-2 hamsters treated as adults, highly efficient gene transfer of -SG also improved muscle morphology (Figure 4a [bottom] and 4c). The CN rates were reversed from >70% before the treatment to <5% after the treatment in hamsters examined at 6 months of age (Figure 4b). Similar reversal was also previously observed after local intramuscular injection of an AAV -SG vector in the hind-leg muscles of the adult Bio14.6 hamsters,14 the parental line of the TO-2 hamsters. The above data demonstrated that in both neonatal and adult TO-2 hamsters, AAV8 vectors could achieve significant therapeutic efficacy in the skeletal muscle by systemic gene transfer.

    Amelioration of Cardiomyopathy and Improvement in Cardiac Functions by Systemic Gene Transfer

    Because congestive heart failure is the major cause of premature death of the TO-2 hamsters and a population of LGMD patients, we next investigated whether robust gene transfer and expression in the hearts could result in efficient therapeutic effects on the cardiomyopathy. Indeed, the treated hamsters, when examined at 8.5 months after neonatal gene therapy, were found to have normal gross heart morphology (Figure 5a, top). The hearts also exhibited normal ventricle structures and lacked any pathological signs of cardiomyopathy, as revealed by hematoxylin and eosin staining of cross sections of the entire heart (Figure 5a, bottom). The morphology and histology were indistinguishable from the age-matched wild-type F1B controls. In contrast, autopsy of the age-matched untreated TO-2 hamsters showed markedly dilated heart, prominent calcification plaques on the heart surface (Figure 5a, top), atrium thrombosis, and secondary pathology of heart failure such as renal dysfunctions, enlarged liver, swollen lungs, and severe ascites in both the chest and peritoneal cavities (data not shown). Microscopic findings indicated cardiomyocyte degeneration and necrosis, pronounced fibrosis, and calcification in the untreated hamsters (Figure 5b). However, those pathological signs were essentially absent in the AAV-treated hamsters (Figure 5b).

    In addition to the histopathology, we also examined the major cardiac functional indices of the hamsters using noninvasive echocardiography.10 The data were collected in hamsters 4 and 8 months of age for the groups treated as neonates and in hamsters 5 months of age for those treated as adults. Significant improvement (Figure 6a and 6b) was observed in percent fractional shortening, an index of systolic function; left ventricle end-systolic and end-diastolic dimensions, indices of chamber dilation; and left ventricle posterior wall thickness, an index of cardiac hypertrophy. In all of these parameters, the treated TO-2 hamsters were statistically equivalent to the wild-type F1B hamsters but significantly different from those of the untreated TO-2 hamsters. These results suggest that systemic delivery of human -SG by the AAV8 vector effectively ameliorated the cardiomyopathy and preserved cardiac structure and function in the TO-2 hamster.

    Improvement in Whole-Body Endurance and Prolongation of Lifespan by Systemic Gene Transfer

    Finally, we examined whether the efficient systemic -SG gene transfer could render whole-body functional rescue and ultimately prolongation of lifespan of the diseased animals. Treadmill running was used to test the whole-body endurance as an indicator of both skeletal and cardiac muscle functions at the age of 4 and 9 months. The running distances (in meters) of the AAV-treated TO-2 hamsters were found to be equivalent to that of normal F1B hamsters but markedly longer than that of untreated TO-2 hamsters in both age groups (AAV8-treated versus untreated TO-2, 765±127 versus 220±46 m at 4 months and 474±113 versus 78±11 m at 9 months) (Figure 7a and video material in the Data Supplement, which is available online at http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.105.565598/DC1). Previously, we have observed in an in vitro test full contractile force recovery as a result of local injection of an AAV-SG vector into the TA muscles of the Bio14.6 hamsters,14 the parental line of the TO-2 hamsters. In this study, we also attempted to measure the TA muscle contractile force improvement in the TO-2 hamsters after systemic gene delivery. Our preliminary results comparing the TA muscles of wild-type F1B and the untreated TO-2 hamsters, however, showed no statistical difference in the generation of specific force (data not shown). This made it difficult to obtain any meaningful evaluation of this parameter in the TO-2 hamsters. A possible reason may be the lack of pathological hypertrophy of the TA muscles in the TO-2 hamsters.

    Furthermore, long-term -SG expression in muscles throughout the body also led to an improvement in the general health and prolongation of the lifespan of the TO-2 hamsters. Without the gene therapy treatment, all of the animals died of heart failure and muscular dystrophy at 32 to 43 weeks of age (median survival time, 37 weeks). The much-shortened lifespan was a major reason for us to choose the TO-2 hamsters in this study to examine the whole-body therapeutic efficacies of gene therapy. As expected, all of the AAV8-treated TO-2 hamsters survived beyond 48 weeks (the duration of the study) (Figure 7b). These data demonstrated the overall efficiency of systemic gene transfer and its global therapeutic effects in the heart failure and muscular dystrophy animal model.

    Discussion

    In the present study, we have for the first time achieved profound and global therapeutic efficacies in a heart failure and muscular dystrophy animal model by means of AAV8-mediated whole-body striated muscle gene delivery without any physical or pharmaceutical interventions. Importantly, the therapeutic efficacies were obtained in both neonatal and adult animals as a result of widespread and extensive transgene transfer and expression. To treat the hereditary muscle diseases, it is vital to deliver the therapeutic genes into as many target cells as possible because the gene products are often cellular proteins with functions that are "cell autologous," unlike circulating proteins such as the clotting factors41 that need only a small percentage of normal levels to obtain significant therapeutic benefits. In this respect, gene therapy and cell therapy for muscular dystrophies are particularly challenging. In our study, the nearly complete gene transfer and long-term, muscle-specific expression of the human -SG gene in both heart and muscle were key to the success of global rescue of the disease phenotypes.

    Recently, we found that AAV8 vectors could cross the capillary blood vessel barriers in the skeletal muscle and heart more efficiently than other AAV serotype vectors tested.3 In the present study, we have extensively investigated the therapeutic effects after AAV8-mediated -SG gene delivery in a severe LGMD animal model, the TO-2 hamsters, which show both muscle pathology and dilated congestive heart failure with a much-shortened lifespan. Previously, gene therapy approaches have been used to treat LGMD hamsters and mice but only with local gene delivery in either the skeletal muscle15,29,30,42,43 or the heart.9,10,31 Until now, technical hurdles and poor efficiency in systemic gene delivery in muscle and heart have prevented achievement of any meaningful whole-body therapeutic benefits such as an increase in treadmill running endurance and the prolongation of lifespan. A recent report described the use of AAV6 and high doses of vascular endothelial growth factor (VEGF) to achieve systemic gene delivery in a Duchenne muscular dystrophy animal model, the mdx mice.19 Those mice do not manifest severe phenotypes of muscular dystrophy such as shortened lifespan seen clinically in Duchenne muscular dystrophy patients, however, so it was difficult to evaluate the life-saving effects of systemic gene delivery. In addition, although vascular endothelial growth factor is a potent blood vessel–permeable reagent that facilitated the dissemination of the AAV6 viral particles into the muscle tissues, the short-term and long-term safety of high dose administration of vascular endothelial growth factor remains to be investigated.21 It is apparently advantageous to achieve systemic gene delivery by AAV8 into the striated muscles without the use of additional pharmaceutical reagents.

    AAV8-mediated systemic gene transfer delivers the genes not only to the muscle and heart but also to other unintended tissues, particularly the adult liver, which has the highest copy numbers of the vector DNA. Nonspecific gene transfer and expression may result in unwanted side effects. Because targeted gene transfer with selective infectivity to the muscle and heart is currently unrealistic, a practical measure we took in this study was the use of an SP that conferred tissue-specific -SG gene in both heart and skeletal muscles. Previously, the most commonly used muscle-specific promoter in the gene therapy studies was the muscle CK promoter and its derivatives.19,44–46 However, the shortened muscle CK promoters were not sufficiently active in the heart and diaphragm19 and may show preference to certain muscle fiber types.44 However, the universal gene expression in the striated muscles by the SP promoter37 observed in our study, particularly in the heart and diaphragm, is very meaningful because cardiac and respiratory failures are the main causes of premature death in dystrophic patients. Muscle-specific expression should also lower the potential immune response that could result from transgene expression in the antigen-presenting cells. Indeed, we did not observe any discernable immune response by the TO-2 hamsters to the human -SG protein that was encoded by the AAV vector under the control of the SP promoter. That may partially contribute to the long-term gene expression after gene delivery in the adult TO-2 hamsters, which had mature immune systems at the time of gene delivery.

    In summary, the unprecedented gene delivery efficiency and therapeutic efficacy for both cardiomyopathy and muscular dystrophy demonstrated here in an animal model should pave the way for further preclinical studies in large animal models and eventually in clinical trials for this and other genetic diseases.

    Acknowledgments

    This study is supported by National Institutes of Health grants AR45967 and NS46546 and Natural Science Foundation of China grant 30028017 to Dr Xiao, as well as by National Institutes of Health grant HL–66949 to Dr McTiernan.

    References

    Gussoni E, Soneoka Y, Strickland CD, Buzney EA, Khan MK, Flint AF, Kunkel LM, Mulligan RC. Dystrophin expression in the mdx mouse restored by stem cell transplantation. Nature. 1999; 401: 390–394.

    Harper SQ, Hauser MA, DelloRusso C, Duan D, Crawford RW, Phelps SF, Harper HA, Robinson AS, Engelhardt JF, Brooks SV, Chamberlain JS. Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy. Nat Med. 2002; 8: 253–261.

    Wang Z, Zhu T, Qiao C, Zhou L, Wang B, Zhang J, Chen C, Li J, Xiao X. Adeno-associated virus serotype 8 efficiently delivers genes to muscle and heart. Nat Biotechnol. 2005; 23: 321–328.

    Sampaolesi M, Torrente Y, Innocenzi A, Tonlorenzi R, D’Antona G, Pellegrino MA, Barresi R, Bresolin N, De Angelis MG, Campbell KP, Bottinelli R, Cossu G. Cell therapy of alpha-sarcoglycan null dystrophic mice through intra-arterial delivery of mesoangioblasts. Science. 2003; 301: 487–492.

    Bachrach E, Li S, Perez AL, Schienda J, Liadaki K, Volinski J, Flint A, Chamberlain J, Kunkel LM. Systemic delivery of human microdystrophin to regenerating mouse dystrophic muscle by muscle progenitor cells. Proc Natl Acad Sci U S A. 2004; 101: 3581–3586.

    Greelish JP, Su LT, Lankford EB, Burkman JM, Chen H, Konig SK, Mercier IM, Desjardins PR, Mitchell MA, Zheng XG, Leferovich J, Gao GP, Balice-Gordon RJ, Wilson JM, Stedman HH. Stable restoration of the sarcoglycan complex in dystrophic muscle perfused with histamine and a recombinant adeno-associated viral vector. Nat Med. 1999; 5: 439–443.

    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.

    Fisher KJ, Jooss K, Alston J, Yang Y, Haecker SE, High K, Pathak R, Raper SE, Wilson JM. Recombinant adeno-associated virus for muscle directed gene therapy. Nat Med. 1997; 3: 306–312.

    Li J, Wang D, Qian S, Chen Z, Zhu T, Xiao X. Efficient and long-term intracardiac gene transfer in delta-sarcoglycan-deficiency hamster by adeno-associated virus-2 vectors. Gene Ther. 2003; 10: 1807–1813.

    Hoshijima M, Ikeda Y, Iwanaga Y, Minamisawa S, Date MO, Gu Y, Iwatate M, Li M, Wang L, Wilson JM, Wang Y, Ross J, Chien KR. Chronic suppression of heart-failure progression by a pseudophosphorylated mutant of phospholamban via in vivo cardiac rAAV gene delivery. Nat Med. 2002; 8: 864–871.

    Chao H, Liu Y, Rabinowitz J, Li C, Samulski RJ, Walsh CE. Several log increase in therapeutic transgene delivery by distinct adeno-associated viral serotype vectors. Mol Ther. 2000; 2: 619–623.

    Goyenvalle A, Vulin A, Fougerousse F, Leturcq F, Kaplan JC, Garcia L, Danos O. Rescue of dystrophic muscle through U7 snRNA-mediated exon skipping. Science. 2004; 306: 1796–1799.

    Wang B, Li J, Xiao X. Adeno-associated virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proc Natl Acad Sci U S A. 2000; 97: 13714–13719.

    Xiao X, Li J, Tsao YP, Dressman D, Hoffman EP, Watchko JF. Full functional rescue of a complete muscle (TA) in dystrophic hamsters by adeno-associated virus vector-directed gene therapy. J Virol. 2000; 74: 1436–1442.

    Cordier L, Hack AA, Scott MO, Barton-Davis ER, Gao G, Wilson JM, McNally EM, Sweeney HL. Rescue of skeletal muscles of gamma-sarcoglycan-deficient mice with adeno-associated virus-mediated gene transfer. Mol Ther. 2000; 1: 119–129.

    Watchko J, O’Day T, Wang B, Zhou L, Tang Y, Li J, Xiao X. Adeno-associated virus vector-mediated minidystrophin gene therapy improves dystrophic muscle contractile function in mdx mice. Hum Gene Ther. 2002; 13: 1451–1460.

    Budker V, Zhang G, Danko I, Williams P, Wolff J. The efficient expression of intravascularly delivered DNA in rat muscle. Gene Ther. 1998; 5: 272–276.

    Hagstrom JE, Hegge J, Zhang G, Noble M, Budker V, Lewis DL, Herweijer H, Wolff JA. A facile nonviral method for delivering genes and siRNAs to skeletal muscle of mammalian limbs. Mol Ther. 2004; 10: 386–398.

    Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, Russell DW, Chamberlain JS. Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nat Med. 2004; 10: 828–834.

    Arruda VR, Stedman HH, Nichols TC, Haskins ME, Nicholson M, Herzog RW, Couto LB, High KA. Regional intravascular delivery of AAV-2-F.IX to skeletal muscle achieves long-term correction of hemophilia B in a large animal model. Blood. 2005; 109: 3458–3464.

    Lee CG, Link H, Baluk P, Homer RJ, Chapoval S, Bhandari V, Kang MJ, Cohn L, Kim YK, McDonald DM, Elias JA. Vascular endothelial growth factor (VEGF) induces remodeling and enhances T(H)2-mediated sensitization and inflammation in the lung. Nat Med. 2004; 10: 1095–1103.

    Gao GP, Alvira MR, Wang L, Calcedo R, Johnston J, Wilson JM. Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy. Proc Natl Acad Sci U S A. 2002; 99: 11854–11859.

    Xiao W, Chirmule N, Berta SC, McCullough B, Gao G, Wilson JM. Gene therapy vectors based on adeno-associated virus type 1. J Virol. 1999; 73: 3994–4003.

    Gao G, Vandenberghe LH, Alvira MR, Lu Y, Calcedo R, Zhou X, Wilson JM. Clades of adeno-associated viruses are widely disseminated in human tissues. J Virol. 2004; 78: 6381–6388.

    Rutledge EA, Halbert CL, Russell DW. Infectious clones and vectors derived from adeno-associated virus (AAV) serotypes other than AAV type 2. J Virol. 1998; 72: 309–319.

    Chiorini JA, Kim F, Yang L, Kotin RM. Cloning and characterization of adeno-associated virus type 5. J Virol. 1999; 73: 1309–1319.

    Chiorini JA, Yang L, Liu Y, Safer B, Kotin RM. Cloning of adeno-associated virus type 4 (AAV4) and generation of recombinant AAV4 particles. J Virol. 1997; 71: 6823–6833.

    Homburger F, Nixon CW, Eppenberger M, Baker JR. Hereditary myopathy in the Syrian hamster: studies on pathogenesis. Ann N Y Acad Sci. 1966; 138: 14–27.

    Holt KH, Lim LE, Straub V, Venzke DP, Duclos F, Anderson RD, Davidson BL, Campbell KP. Functional rescue of the sarcoglycan complex in the BIO 14.6 hamster using delta-sarcoglycan gene transfer. Mol Cell. 1998; 1: 841–848.

    Li J, Dressman D, Tsao YP, Sakamoto A, Hoffman EP, Xiao X. rAAV vector-mediated sarcogylcan gene transfer in a hamster model for limb girdle muscular dystrophy. Gene Ther. 1999; 6: 74–82.

    Ikeda Y, Gu Y, Iwanaga Y, Hoshijima M, Oh SS, Giordano FJ, Chen J, Nigro V, Peterson KL, Chien KR, Ross J Jr. Restoration of deficient membrane proteins in the cardiomyopathic hamster by in vivo cardiac gene transfer. Circulation. 2002; 105: 502–508.

    Yue Y, Li Z, Harper SQ, Davisson RL, Chamberlain JS, Duan D. Microdystrophin gene therapy of cardiomyopathy restores dystrophin-glycoprotein complex and improves sarcolemma integrity in the mdx mouse heart. Circulation. 2003; 108: 1626–1632.

    Nigro V, Okazaki Y, Belsito A, Piluso G, Matsuda Y, Politano L, Nigro G, Ventura C, Abbondanza C, Molinari AM, Acampora D, Nishimura M, Hayashizaki Y, Puca GA. Identification of the Syrian hamster cardiomyopathy gene. Hum Mol Genet. 1997; 6: 601–607.

    Sakamoto A, Ono K, Abe M, Jasmin G, Eki T, Murakami Y, Masaki T, Toyo-oka T, Hanaoka F. Both hypertrophic and dilated cardiomyopathies are caused by mutation of the same gene, delta-sarcoglycan, in hamster: an animal model of disrupted dystrophin-associated glycoprotein complex. Proc Natl Acad Sci U S A. 1997; 94: 13873–13878.

    Hoffman EP. Counting muscular dystrophies in the post-molecular census. J Neurol Sci. 1999; 164: 3–6.

    Lim LE, Campbell KP. The sarcoglycan complex in limb-girdle muscular dystrophy. Curr Opin Neurol. 1998; 11: 443–452.

    Li X, Eastman EM, Schwartz RJ, Draghia-Akli R. Synthetic muscle promoters: activities exceeding naturally occurring regulatory sequences. Nat Biotechnol. 1999; 17: 241–245.

    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.

    Kadokami T, McTiernan CF, Kubota T, Frye CS, Bounoutas GS, Robbins PD, Watkins SC, Feldman AM. Effects of soluble TNF receptor treatment on lipopolysaccharide-induced myocardial cytokine expression. Am J Physiol Heart Circ Physiol. 2001; 280: H2281–H2291.

    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.

    Kay MA, High K. Gene therapy for the hemophilias. Proc Natl Acad Sci U S A. 1999; 96: 9973–9975.

    Xiao W, Chirmule N, Schnell MA, Tazelaar J, Hughes JV, Wilson JM. Route of administration determines induction of T-cell-independent humoral responses to adeno-associated virus vectors. Mol Ther. 2000; 1: 323–329.

    Dressman D, Araishi K, Imamura M, Sasaoka T, Liu LA, Engvall E, Hoffman EP. Delivery of alpha- and beta-sarcoglycan by recombinant adeno-associated virus: efficient rescue of muscle, but differential toxicity. Hum Gene Ther. 2002; 13: 1631–1646.

    Dunant P, Larochelle N, Thirion C, Stucka R, Ursu D, Petrof BJ, Wolf E, Lochmuller H. Expression of dystrophin driven by the 1.35-kb MCK promoter ameliorates muscular dystrophy in fast, but not in slow muscles of transgenic mdx mice. Mol Ther. 2003; 8: 80–89.

    Larochelle N, Lochmuller H, Zhao J, Jani A, Hallauer P, Hastings KE, Massie B, Prescott S, Petrof BJ, Karpati G, Nalbantoglu J. Efficient muscle-specific transgene expression after adenovirus- mediated gene transfer in mice using a 1.35 kb muscle creatine kinase promoter/enhancer. Gene Ther. 1997; 4: 465–472.

    Jaynes JB, Johnson JE, Buskin JN, Gartside CL, Hauschka SD. The muscle creatine kinase gene is regulated by multiple upstream elements, including a muscle-specific enhancer. Mol Cell Biol. 1988; 8: 62–70.(Tong Zhu, MD, PhD; Liqiao)