Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: Expert Opin Orphan Drugs. 2015 Dec 17;4(2):169–183. doi: 10.1517/21678707.2016.1124039

Prospect of gene therapy for cardiomyopathy in hereditary muscular dystrophy

Yongping Yue 1, Ibrahim M Binalsheikh 2, Stacey B Leach 3, Timothy L Domeier 4, Dongsheng Duan 1,5,*
PMCID: PMC4914135  NIHMSID: NIHMS794732  PMID: 27340611

Abstract

Introduction

Cardiac involvement is a common feature in muscular dystrophies. It presents as heart failure and/or arrhythmia. Traditionally, dystrophic cardiomyopathy is treated with symptom-relieving medications. Identification of disease-causing genes and investigation on pathogenic mechanisms have opened new opportunities to treat dystrophic cardiomyopathy with gene therapy. Replacing/repairing the mutated gene and/or targeting the pathogenic process/mechanisms using alternative genes may attenuate heart disease in muscular dystrophies.

Areas covered

Duchenne muscular dystrophy is the most common muscular dystrophy. Duchenne cardiomyopathy has been the primary focus of ongoing dystrophic cardiomyopathy gene therapy studies. Here, we use Duchenne cardiomyopathy gene therapy to showcase recent developments and to outline the path forward. We also discuss gene therapy status for cardiomyopathy associated with limb-girdle and congenital muscular dystrophies, and myotonic dystrophy.

Expert opinion

Gene therapy for dystrophic cardiomyopathy has taken a slow but steady path forward. Preclinical studies over the last decades have addressed many fundamental questions. Adeno-associated virus-mediated gene therapy has significantly improved the outcomes in rodent models of Duchenne and limb girdle muscular dystrophies. Validation of these encouraging results in large animal models will pave the way to future human trials.

Keywords: AAV, adeno-associated virus, gene therapy, cardiomyopathy, Duchenne muscular dystrophy, DMD, dystrophin, capsid engineering, vector, capsid, muscular dystrophy, gene therapy, heart failure, limb-girdle muscular dystrophy, congenital muscular dystrophy, myotonic dystrophy

1. Clinical presentation, pathogenic mechanism and therapeutic challenge of dystrophic cardiomyopathy

Dystrophic cardiomyopathy refers to cardiac manifestations of muscular dystrophies. Muscular dystrophies are a clinically, genetically, and biochemically heterogeneous group of disorders. They are characterized by progressive muscle wasting, force loss and dystrophic muscle pathology1, 2. Muscular dystrophies can be classified in many different ways such as the age of onset (congenital/neonatal, adolescent, or adult), disease progression (rapid or slow), the muscle groups involved (such as limb girdle, facioscapulohumeral and oculopharyngeal etc.), the mode of inheritance (such as X-linked/autosomal, recessive/dominant). Some muscular dystrophies are named after people who discovered the disease (such as Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and Emery-Dreifuss muscular dystrophy etc). Despite the unique clinical features of each type of muscular dystrophy, cardiac involvement has been a common finding in most muscular dystrophies and often represents a major cause of morbidity and mortality37. Interestingly, the cardiac phenotype varies in different types of muscular dystrophies and even in different patients or disease stages of the same type muscular dystrophy. Some present with dilated/hypertrophic/restrictive cardiomyopathy with eventually heart failure while others exhibit conduction defects leading to arrhythmia and sudden cardiac death. In the case of DMD and BMD, MRI studies have revealed a unique pattern of subepicardial fibrosis predominantly in the left ventricular lateral wall810.

The pathogenic mechanisms of dystrophic cardiomyopathy are not completely understood11, 12. However, it may at least involve destabilization of the cardiomyocyte membrane, or sarcolemma. Unlike other cells in the body, muscle cells undergo continuous calcium-regulated contraction/relaxation cycles. A consequence of this unique physiology is the repeated cycles of shrinking and expansion of the cell. This dynamic deformation process places enormous stress on the sarcolemma. Such stress is especially problematic for cardiomyocytes because of the repetitive pumping activity of the heart. To relieve contraction-induced stress, muscle cells have evolved specialized trans-membrane protein complexes such as the dystrophin-associated glycoprotein complex (DGC) and the integrin complex. These protein complexes constitute physical connections between the cytoskeleton and the extracellular matrix. Mutations in the genes encoding the components of these complexes result in various forms of muscular dystrophies. Failure to maintain sarcolemmal integrity leads to membrane leakage, myocyte degeneration, necrosis and eventual replacement by fibrofatty tissue. Clearly, strengthening the destabilized sarcolemma holds the key for treating dystrophic cardiomyopathy. Unfortunately, this cannot be achieved with conventional medical/surgical treatments13. Gene therapy, however, provides a great opportunity to address this therapeutic challenge.

2. Strategies to deliver a therapeutic gene to a dystrophic heart

Disease-causing genes for many muscular dystrophies have been discovered. The identification of the genetic underpinning makes it possible to treat dystrophic cardiomyopathy with gene therapy. The first step of gene therapy is delivery of a therapeutic gene to the heart. A number of viral and nonviral vectors have been tested14. So far the most effective and least immunogenic vector is adeno-associated virus (AAV). AAV is a 20-nm single stranded DNA virus15. Recombinant AAV vector contains no wild type viral genes. The vector genome can be readily packaged into naturally existing or synthetic capsids to meet specific therapeutic needs. The nano-size AAV particle creates a packaging dilemma. The maximal carrying capacity of a single AAV particle is 5-kb16. This is too small for many genes required for muscular dystrophy gene therapy (such as the dystrophin gene and the dysferlin gene). To overcome this limitation, we and others have invented a series of dual and tri-AAV vectors17. The basic idea is to fragment a large therapeutic gene and package each segment into an AAV particle. The full-length gene is reconstituted by cellular recombination machinery after co-infection. These multi-vector strategies have made it possible to deliver the 6 to 8-kb mini-dystrophin gene and even the 12-kb full-length dystrophin coding sequence to dystrophin-deficient mdx mice, the most commonly used animal models for DMD1822.

Over the years, a number of different strategies have been developed to achieve effective AAV gene transfer in dystrophic hearts. Early studies were mainly based on AAV-2 using invasive and complicated methods such as direct myocardial injection23, intracavity injection24, transcoronary perfusion25, and ex vivo coronary perfusion26. The identification and development of novel AAV capsids has opened the door to transduce dystrophic hearts with peripheral vein injections2732. This simple method not only greatly reduces the risks associated invasive heart gene transfer but also allows simultaneous treatment of both cardiac and skeletal muscle disease in muscular dystrophy.

The tissue tropism of the AAV vector is largely determined by the viral capsids. Experimenting with natural and engineered AAV capsids has proven to be a fruitful approach in identifying cardiotropic AAV vectors. For example, a comparison of AAV-1 to 9 revealed AAV-9 as the most potent vector for the mouse heart33. Indeed, AAV-9 results in robust widespread myocardial transduction in mdx mice irrespective of the age and the route of delivery (intravenous or intra-arterial)3436. Directed evolution and cardiotropic peptide insertion have also yielded novel AAV variants with enhanced cardiac transduction in rodent models of limb girdle muscular dystrophy (LGMD) 2F, an extremely rare type of muscular dystrophy caused by δ-sarcoglycan deficiency32, 37, 38.

3. Disease gene-specific gene therapy

3.1. Dystrophin-based Duchenne cardiomyopathy gene therapy

The dystrophin gene was the first muscular dystrophy-associated gene cloned39. Its mutation leads to DMD. The 2.4-mb full-length dystrophin gene contains 79 exons and it transcribes into a ~ 12-kb cDNA. The full-length dystrophin protein has four major functional domains including the N-terminal, rod, cysteine-rich and C-terminal domain. The N-terminal domain binds to cytosolic γ-actin. The rod domain consists of 24 spectrin-like repeats. Within the rod domain, there are several important subdomains including one for γ-actin-binding, one for neuronal nitric oxide synthase (nNOS)-binding and one for microtubule-binding4043. The cysteine-rich domain links dystrophin to the extracellular matrix through dystroglycan, a transmembrane glycoprotein. The C-terminal domain binds to syntrophin and dystrobrevin.

The enormous size of the dystrophin gene presents a delivery challenge because it is beyond the packaging capacity of most viral vectors. Interestingly, some naturally-existing, internally-deleted dystrophins (e.g. Δ17–48) are quite functional44. These mini-dystrophin genes are about 6 to 8-kb in length and their expression in humans and animals has greatly mitigated skeletal muscle disease1921, 4447. The therapeutic implication of mini-dystrophin in the heart has only been investigated in transgenic mice48. We expressed mini-dystrophin specifically in the heart of mdx mice. This cardiac-restricted expression completely corrected cardiac histopathology, improved exercise performance and enhanced myocardial contractility48. Whether mini-dystrophin gene therapy can achieve similar effectiveness remains to be seen. In this regard, dual AAV vectors have been developed to express the mini-dystrophin gene1821, 4951. Further, systemic injection of dual AAV vectors has been shown to transduce the myocardium at high efficiency in mdx mice52, 53.

A single vector therapy would be more advantageous. To package dystrophin into AAV, highly abbreviated micro-dystrophin genes have been developed. The microgene is about 3.5 to 4-kb in length and contains ~30% of the dystrophin coding sequence. In contrast to mini-dystrophin, micro-dystrophin does not carry the C-terminal domain. Additionally, it has a shorter rod domain with only 4 to 5 spectrin-like repeats. AAV-mediated micro-dystrophin gene therapy has been extensively studied in various mouse models and more recently in the canine model47, 5459. Direct or systemic AAV microgene therapy significantly ameliorated skeletal muscle disease in dystrophic mice and dogs. The first study to evaluate therapeutic effect of micro-dystrophin in the heart was performed by Yue et al24. In this study, an AAV-5 microgene vector was directly injected into the cardiac cavity of neonatal mdx mice. Micro-dystrophin restored the DGC complex in the heart and enhanced the membrane stability of cardiomyocytes24. In subsequent studies, newly developed AAV capsids (such as AAV-6 and AAV-9) were utilized to delivery micro-dystrophin to the heart through peripheral vein injection3436, 54, 6062. Of particular interest are studies by Bostick et al in which an AAV-9 microgene vector was delivered to the heart of aged female mdx mice. This study is noteworthy because aged female mdx mice develop a cardiac phenotype nearly identical to that observed in dilated cardiomyopathy of human patients34, 35, 63, 64. Despite the advanced heart disease in very old mice, surprisingly, cardiomyocytes were efficiently transduced34, 35. The average lifespan of mdx mice is ~ 22 months65, 66. In pre-terminal mdx mice (16 to 20-m-old), microgene therapy reduced myocardial fibrosis, improved the electrocardiographic profile and hemodynamic function34. In terminal age mdx mice (> 21-m-old), neither fibrosis nor hemodynamic function was improved35. However, some ECG parameters were partially corrected and dobutamine stress-induced acute cardiac death was reduced35.

Expression of a full-length or near-full-length dystrophin protein may lead to a better recovery. This is feasible with tri-AAV vectors but the efficiency is too low to be of practical use22. Editing the mutated RNA transcript or genome offers alternative approaches to reach this goal. Exon skipping is a potent method to achieve RNA-level editing. Briefly, antisense oligonucleotides (AONs) are delivered to modulate RNA splicing so that the mutated (and some times adjacent) exons are removed. The resulting mRNA, though abbreviated, is in-frame and can yield a near-full-length protein67. Several chemically distinctive classes of AONs have been developed including 2-O-methylated phosphorothioated (2-OMePS), phosphorodiamidate morpholino oligomers (PMOs), peptide/polymer/nanoparticle–conjugated PMOs, and most recently tricycle-DNA (tcDNA). 2-OMePS and PMOs are currently in clinical trials6873. However, these AONs cannot reach the heart7476. Peptide/polymer/nanoparticle–conjugated PMOs can induce exon-skipping in the heart of mdx mice and improve heart function7787. However, there are issues related to potential toxicity and immunogenicity88. The newly developed tcDNA represents the most advanced AON formulation89. Because of its unique pharmacological property, systemic delivery of tcDNA-AONs resulted in phenomenal uptake in many tissues including the heart and brain. Treatment in mdx mice and more severe utrophin/dystrophin double knockout (u-dko) mice improved cardiac, respiratory and behavioral function89. Importantly, no overt toxicity was detected with tcDNA89. An alternative strategy to deliver AONs is to use the AAV vector. AAV-9 mediated systemic AON delivery resulted in high efficient dystrophin expression in the heart of u-dko mice90. More recently, two independent groups achieved long-term dystrophin restoration in the heart of the canine DMD model with AAV-6 mediated local exon-skipping91, 92.

Compared to RNA editing with exon-skipping, targeted editing of the mutated dystrophin gene has just entered an exciting time due to recent development of highly versatile genome engineering tools such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and most importantly, the clustered regularly interspaced palindromic repeat (CRISPR)-associated endonuclease 9 (Cas9)93. A series of elegant studies from the Gersbach laboratory has provided compelling proof-of-concept evidence in correcting cells from DMD patients using these new technologies9496. It is highly anticipated that genome editing will soon be used to treat skeletal muscle disease and cardiomyopathy in animal models of DMD97.

3.2. Disease gene-based therapy for cardiomyopathy in other muscular dystrophies

3.2.1. Targeting disease gene to treat LGMD cardiomyopathy

LGMD refers to a group of muscle disorders with a wide range of clinical and genetic heterogeneity98, 99. Based on the inheritance pattern, they are classified as autosomal dominant type 1 (LGMD1) and autosomal recessive type 2 (LGMD2). Each type of LGMD is further classified according to the time the disease gene was discovered. For LGMD1, the goal of the gene therapy is to decrease the expression of the mutated gene. This can be achieved with RNA interference (RNAi) to silence the mutated gene100, 101. So far, only one study has tested gene therapy for dominant LGMD. LGMD1A is caused by myotilin gene mutation. Liu et al targeted mutant myotilin with an AAV-6 microRNA vector102. Treatment significantly reduced expression of the mutated myotilin protein and ameliorated skeletal muscle myopathy. Although LGMD1A patients do not exhibit cardiac abnormalities4, the RNAi approach described by Liu et al may treat cardiac manifestations in other dominant myopathies such as lamin A/C gene mutation-induced LGMD1B and Emery-Dreifuss muscular dystrophy.

There has been significant progress in LGMD2 gene therapy. AAV-mediated gene therapy has been tested in animal models of at least seven different subtypes of LGMD2 (2A to 2F, and 2I). LGMD2A and 2B are caused by mutations in the calpain-3 gene and the dysferlin gene, respectively. According to Hermans, LGMD2A and 2B do not show cardiac manifestations4. However, cardiomyopathy has been seen in dysferlin-deficient mice and there are also a few reports of cardiac involvement in some LGMD2B patients103106. Three different approaches have been explored to express a function dysferlin gene. These include delivering a minimized dysferlin gene with a single AAV vector, delivering a full-length dysferlin cDNA with dual AAV vectors and exon-skipping or pre-mRNA trans-splicing to repair defective dysferlin RNA transcript107113. Defective membrane repair has been considered as the major pathogenic mechanism for LGMD2B. The in vitro membrane repair assay has been used as a surrogate endpoint to evaluate the therapeutic efficacy. Surprisingly, a recent study by Lostal et al found that a correction of membrane repair by the in vitro assay did not correlate with the correction of muscle pathology. The authors overexpressed myoferlin, a homolog of dysferlin, in dysferlin-null mice by the transgenic approach and they also expressed the mini-dysferlin gene in 4-week-old dysferlin-null mice. Neither transgenic overexpression of myoferlin nor AAV-mediated expression of mini-dysferlin improved muscle histology although both corrected membrane repair deficit in vitro114.

LGMD2C to 2F are often referred to as sarcoglycanopathies because they are caused by mutations in the sarcoglycan genes. The most common sarcoglycanopathy is α-sarcoglycan-deficient LGMD2D. Cardiac involvement is rare in LGMD2D6. LGMD2C, which is caused by mutations in the γ-sarcoglycan gene, usually exhibits mild cardiomyopathy. Deficiency of β-sarcoglycan and δ-sarcoglycan results in LGMD2E and LGMD2F, respectively. These two diseases are associated with dilated cardiomyopathy3, 6. The molecular weights of sarcoglycans are 35 to 50 kD. The small size makes sarcoglycan genes perfect candidates for AAV delivery. Sarcoglycanopathies were among the first few inherited diseases proposed for AAV gene therapy115. Recently, AAV gene therapy for LGMD2C and 2D has entered into clinical trials116118. Very few studies have explored AAV β-sarcoglycan gene transfer for treating LGMD2E119, 120. However, therapeutic delivery of the δ-sarcoglycan gene by AAV has been tested extensively in the mouse and hamster models of LGMD2F. Systemic or direct myocardial delivery of the δ-sarcoglycan gene not only reduced histological lesions in the heart (such as myocardial necrosis, inflammation, calcification and fibrosis) but also improved heart function and extended lifespan38, 121125. Collectively, these preclinical studies suggest that AAV δ-sarcoglycan gene transfer is an effective treatment for dilated cardiomyopathy in LGMD2F.

LGMD2I is caused by mutations in the fukutin-related protein (FKRP) gene. FKRP is located in the Golgi apparatus and it is essential for post-translational glycosylation of α-dystroglycan, the protein that directly interacts with the extracellular matrix in the DGC complex. More than half of LGMD2I patients have cardiac abnormalities and a quarter of them develop heart failure126. Gene therapy for LGMD2I has been hindered by the lack of a good animal model. Nonsense mutations and whole gene deletions are embryonically lethal127. To overcome this hurdle, Lu and colleagues recently generated a FKRP L276I knock-in mouse128. This nonsense mutation model mimics the clinical phenotype of LGMD2I. To determine whether systemic delivery of the FKRP gene with AAV can protect the heart, Qiao et al performed intraperitoneal injection in newborn FKRP L276I knock-in mice using an AAV-8 vector. Dobutamine-stressed echocardiography in 7-m-old treated mice showed significantly higher ejection fraction and fractional shortening than those of untreated mice128.

3.2.2. Targeting disease gene to treat heart disease in other muscular dystrophies

Dystroglycanopathies are a group of congenital muscular dystrophies (MDC). They are caused by mutations in the genes involved in the glycosylation pathway of α-dystroglycan129, 130. Fukuyama muscular dystrophy, a dystroglycanopathy caused by retrotransposon insertion in the 3′-untranslated region of the fukutin gene, is associated with severe cardiomyopathy and congestive heart failure131, 132. Blockade of pathogenic exon-trapping by a cocktail of AONs restored fukutin expression and α-dystroglycan glycosylation in the mouse model and human cells132. Whether this therapy can rescue heart function remains to be determined by future studies.

FRKP gene mutation not only causes LGMD2I but also causes congenital muscular dystrophy type 1C (MDC1C). Similar to LGMD2I, cardiac involvement is also a frequent finding in MDC1C patients133. A mouse model for MDC1C has been generated with FKRP P448L knock-in134. AAV-9 mediated FKRP expression normalized α-dystroglycan glycosylation in the heart of MDC1C mice. Unfortunately, cardiac function was not assessed due to mild heart disease at the age of euthanization (5 months)134.

Myotonic dystrophy (DM), the second most common muscular dystrophy, is an autosomal dominant disease. It is caused by pathogenic RNA gain-of-function toxicity due to CTG (for DM1) or CCTG (for DM2) expansion. Cardiac conduction deficits (conduction block and arrhythmia) contribute significantly to the morbidity and mortality135. About 20 different mouse models have been developed to reveal various aspects of the disease136. Among these, tamoxifen-inducible EpA960 mice and tetracycline-inducible GFP-DMPK-(CTG)5 mice are considered as good models to test cardiac interventions for DM137, 138. The field of DM gene therapy has been particularly active in recent years. RNAi, ribozyme, AONs and more recently site-specific RNA endonuclease have all been explored for DM gene therapy139144. However, most of these studies have not examined therapeutic efficacy in the heart. The in vivo proof of principle for reversing cardiac conduction defects has only been shown in GFP-DMPK-(CTG)5 mice. In this model, administration of doxycycline induced myotonia and cardiac conduction abnormalities. Discontinuation of doxycycline dramatically reduced myotonic symptoms and conduction block in the heart137.

4. Expanding the armory of dystrophic cardiomyopathy gene therapy by targeting pathogenic mechanisms

4.1. Dystrophin-independent Duchenne cardiomyopathy gene therapy

4.1.1. Stabilization of cardiomyocyte membrane with endogenous cellular genes

Given membrane weakening is a primary pathogenic mechanism, strategies that enhance sarcolemmal stability should theoretically ameliorate Duchenne cardiomyopathy. Utrophin is a dystrophin homolog145. Despite some differences43, 146, 147, utrophin shares significant structural and functional similarity to dystrophin and assembles the utrophin-associated glycoprotein complex (UGC). As is the case for dystrophin, micro-utrophin has been generated for AAV delivery40, 148. More recently, AAV-mediated expression of jazz, an artificial zinc finger transcription factor, was found to activate the utrophin promoter and enhance utrophin expression149. So far these utrophin-based strategies have only been shown to protect skeletal muscle. Their therapeutic efficacy in the heart remains to be tested experimentally. Several components of the DGC and UGC, including sarcoglycans, sarcospan and nNOS, were recently shown to reduce the skeletal muscle phenotype in mdx mice66, 150, 151. Of these, only nNOS has been shown to treat Duchenne cardiomyopathy152. Specifically, Lai et al delivered a PDZ domain truncated version of the nNOS gene to the heart of 14-m-old mdx mice and examined the cardiac phenotype when mice reached 21 months of age. Supra-physiological ΔPDZ-nNOS expression significantly reduced myocardial fibrosis, inflammation and apoptosis. Importantly, treatment partially ameliorated ECG abnormalities and improved hemodynamic performance152.

Besides the DGC and UGC, the integrin complex (especially α7β1) is another membrane-crossing complex that stabilizes the sarcolemma153. Expression of the α7-integrin gene by AAV was recently shown to reduce limb muscle disease in mdx mice and extend the life span of u-dko mice154, 155. The cardiac benefit of AAV-mediated α7-integrin expression remains to be demonstrated.

4.1.2. Treating Duchenne cardiomyopathy with calcium regulating genes

Cytosolic calcium overload is a pivotal pathogenic event leading to muscle damage and force reduction in DMD156. Restoring calcium homeostasis holds great promise for treating Duchenne cardiomyopathy. The sarco/endoplasmic reticulum calcium ATPase (SERCA) is a calcium pump that removes calcium from the cytosol and transports it into the lumen of the sarcoplasmic reticulum (SR). SERCA accounts for ≥ 70% of calcium removal from the cytosol in muscle cells. SERCA2a is expressed in the heart and slow twitch skeletal muscle. We found SERCA2a expression is reduced in the heart of mdx mice by immunostaining157. When the AAV-9 SERCA2a vector was delivered to the heart of 12-m-old mdx mice, it increased myocardial SERCA2a expression and significantly improved cardiac electrophysiology157. Encouragingly, similar protection was observed when the AAV-9 SERCA2a vector was administrated to terminal aged (22-m-old) mdx mice158.

4.1.3. Additional dystrophin-independent gene therapy strategies

Besides strengthening the sarcolemma and restoring calcium homeostasis, investigators have explored many other creative gene therapy strategies that are not dependent on dystrophin. These include AAV-mediated inhibition of the myostatin pathway, AAV-mediated overexpression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), the cytotoxic T cell GalNAc transferase (Galgt2) and miR486, and AAV-mediated blocking of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway159165. However, most of these studies only demonstrated disease amelioration in skeletal muscle. Whether cardiac muscle can be protected is yet to be seen. Among these strategies, the myostatin inhibition approach is especially intriguing because this approach aims at increasing muscle mass. This raises two concerns: (a) muscle hypertrophy may increase stress on the sarcolemma and hence worsen muscle disease, and (b) myostatin inhibition may lead to hypertrophic cardiomyopathy. Indeed, different results have been achieved depending on the models used. In animal models for DMD (mice and dogs), myostatin inhibition has consistently improved skeletal muscle pathology and function163, 164, 166168. In a phase I trial, AAV-mediated regional expression of the myostatin antagonist follistatin improved walking distance in 5 out of 6 BMD patients169. However, the results of myostatin inhibition appears less promising in preclinical studies of some other muscular dystrophies such as LGMD2B, LGMD2C, LGMD2F and congenital muscular dystrophy type 1A170173. Cohn et al examined whether myostatin deficiency can cause myocardial hypertrophy in normal C57BL/6 mice and mdx mice174. Surprisingly, myostatin elimination did not affect heart weight and heart weight/body weight ratio in either strain174. A major protective mechanism of myostatin inhibition is to reverse muscle fibrosis by inducing fibroblast apoptosis175. For reasons yet unknown, this mechanism appears to be deficient in the heart174. Collectively, there is a lack of clear evidence suggesting that myostatin blockade benefits a dystrophic heart. Myostatin inhibition-based gene therapy strategies have to be carefully weighted against potential undesirable side effects170173, 176.

4.2. Disease gene-independent gene therapy for cardiomyopathy in other muscular dystrophies

4.2.1. Disease gene independent gene therapy for dilated cardiomyopathy in LGMD2E and 2F

MicroRNAs (miRs) are regulatory non-coding RNAs. Recent studies suggest that miRs play crucial roles in myocardial remodeling177. Sampaolesi and colleagues found that miR669 is down regulated in the heart of β-sarcoglycan null LGMD2E mice178. In a subsequent study, they evaluated preventive miR gene therapy in β-sarcoglycan knockout mice179. After intra-ventricular delivery of an AAV-2 miR669a vector to neonates, they quantified survival, cardiac fibrosis and function at the age of 18 months. AAV injected mice showed significantly better survival, less myocardial fibrosis and better heart function179.

Several disease gene-independent approaches have been tested to treat dilated cardiomyopathy in rodent models of LGMD2F25. Mitsugumin 53 (MG53) is a 53 kD membrane repair protein and also a ubiquitin E3 ligase180. Mice lacking MG53 show increased susceptibility to sarcolemmal injury and develop a slow but progressive myopathy181. He et al introduced MG53 to neonatal and young adult LGMD2F hamster model with AAV-8. Supra-physiological MG53 expression in the heart and limb muscle partially reduced the serum creatine kinase level, stabilized the sarcolemma, and slowed muscle degeneration and fibrosis. It also improved treadmill performance and heart function182. Since sarcolemmal disruption is a common pathogenic process, it is suggested that MG53 therapy may server as a broadband therapeutics for a wide range of muscular dystrophies180. Unfortunately, there are some important safety concerns for long-term use. In one study, authors noticed elevation of hepatic enzymes due to leaky MG53 expression in the liver182. Most alarmingly, two recent studies found that the E3 ligase function of MG53 targets the muscle insulin receptor and insulin-receptor substrate 1 for degradation183, 184. Transgenic over-expression of MG53 in striated muscle and heart resulted in metabolic syndrome and diabetic cardiomyopathy, respectively183, 185.

Defects in sarcoplasmic reticulum calcium cycling plays a pivotal role in the pathogenesis of inherited and acquired cardiomyopathy186. As eluded before, SERCA2a is the primary calcium pump in the heart. AAV-mediated SERCA2a over-expression ameliorates some cardiac manifestations in the mdx model of Duchenne cardiomyopathy157, 158. The activity of SERCA2a is regulated by phospholamban. Unphosphorylated phospholamban inhibits SERCA2a activity but phosphorylated phospholamban does not. A single amino acid change (Ser16 Glu) locks phospholamban in a conformation that resembles the phosphorylated form. Hoshijima et al delivered this pseudo-phosphorylated phospholamban to the heart of δ-sarcoglycan deficient hamsters using AAV-225. Chronic expression of pseudo-phosphorylated phospholamban markedly improved heart function in this LGMD2F dilated cardiomyopathy model25.

Apoptosis has been implicated in the progression of heart failure. In particular, activation of apoptosis signal-regulating kinase 1 (ASK1) induces cardiomyocytes apoptosis. Hikoso et al tested whether delivery of the dominant mutant form of ASK1 can reduce cardiomyopathy in the LGMD2F hamster model187. They delivered dominant mutant ASK1 by AAV-2 via transcoronary perfusion to 10-week-old affected hamsters. Evaluation at the age of 24 weeks revealed remarkable improvements of systolic and diastolic function as well as a reduction of chamber dilation and myocardial fibrosis.

4.2.2. Disease gene independent gene therapy for cardiomyopathy in other muscular dystrophies

Merosin (laminin α2) is an extracellular matrix protein. Deficiency in merosin leads to congenital muscular dystrophy MDC1A. Although MDC1A patients usually do not have clinically significant cardiomyopathy4, cardiac involvement has been documented in atypical patients and laminin α2-null dy/dy mice188191. Agrin is also an extracellular matrix protein but it has no structural similarity to laminin α2. Interestingly, AAV-1 mediated systemic expression of a miniature version of agrin greatly reduced myocardial fibrosis in dy/dy mice192.

LGMD2I and MDC1C are caused by mutations in the FKRP gene and both diseases display prominent cardiac manifestations. FKRP knock-in mice L276I and P448L have been developed to model LGMD2I and MDC1C, respectively128, 134. The pathway of α-dystroglycan glycosylation involves a series of glycosyltransferases. Like-acetylglucosaminyltransferase (LARGE) acts downstream of FKRP. Activation of a downstream enzyme presumably should correct the disease phenotype caused by upstream enzyme deficiency. Vannoy indeed found that AAV-mediated LARGE over-expression not only reduced myopathy in LARGE-deficient congenital muscular dystrophy mice but also improved α-dystroglycan glycosylation in the heart and skeletal muscle of FKRB P448L knock-in mice193.

5. Expert opinion

The cloning of the dystrophin gene in 1986 started a flood of discoveries on genes whose mutations cause various forms of muscular dystrophies39. All of a sudden, it appears we may cure many muscular dystrophies and their associated cardiomyopathy by either fixing the mutated gene or introducing a functional copy of the normal gene. While conceptually straightforward, the journey thus far has turned out to be long and winding. Research in dystrophic cardiomyopathy and its gene therapy has made significant progress in the last decade194196. Several fundamental issues have been addressed. These include the establishment of a large collection of animal models to test experimental gene therapy in various forms of dystrophic cardiomyopathy, the development of noninvasive AAV delivery methods to efficiently transduce the heart, and the expansion of therapeutic schemes from simply delivering a functional cDNA to dystrophic muscle to the modulation of the RNA/DNA structure and expression using a variety of coding and noncoding sequences, even oligonucleotides. Some critical parameters for dystrophic cardiomyopathy gene therapy have also been clarified. For example, studies in the mdx model of Duchenne cardiomyopathy have provided compelling evidence that we may achieve a near wild-type protection by treating half of the cardiomyocytes instead of every single cell63, 197. On the other hand, debates on whether treating skeletal muscle disease will alleviate or aggravate cardiomyopathy have settled down on the conclusion that both should be treated either together or separately76, 198, 199.

There is no doubt that Duchenne cardiomyopathy gene therapy has led the way for the entire field. First, a number of models have been generated for Duchenne cardiomyopathy gene therapy studies such as aged mdx mice, Cmah/mdx mice, u-dko mice, myoD/dystrophin double knockout mice and telomerase RNA/dystrophin double-null mdx/mTR mice63, 64, 200204. Importantly most of these rodent models are commercially available205. In terms of large animal models, besides the commonly used golden retriever muscular dystrophy dogs (GRMDs), additional dog models have been identified and colonies established206209. Second, we have successfully treated the cardiac phenotype in symptomatic u-dko mice and aged mdx mice using micro-dystrophin and exon-skipping34, 89. We even achieved widespread myocardial AAV gene transfer and some ECG improvements in terminal stage mdx mice35. For scaling up, efficient myocardial transduction has been achieved in newborn dogs and adult affected dogs with systemic and percutaneous transendocardial AAV delivery57, 91, 92, 210, 211. Third, many previously under-appreciated disease targets (such as nNOS and SERCA2a) and revolutionary technologies (such as tcDNA, ZENs, TALENs and CRISPR/Cas9) are now on the horizon for Duchenne cardiomyopathy gene therapy. Despite this substantial progress, we still do not have answers to a lot of important questions. For example, it is not clear whether supra-physiological dystrophin expression in the heart is toxic, whether there exists heart-specific domain(s) in the dystrophin gene that should be included in micro-dystrophin, and whether cardiotropic features of some existing AAV serotypes can cross the species boundary and result in efficient heart transduction in humans. For this last point, some recent developments in the generation of the xenograft model using dystrophic human muscle and forced evolution of human tissue tropic AAV capsids may provide some hints32, 212, 213. It should be noted that emerging new technologies such as genome editing with CRISPR/Cas9 not only brings in new hopes, they are also accompanied with new questions such as potential toxicity from off-target editing.

There is a long to-do list for the field of dystrophic cardiomyopathy gene therapy. Some of these may include (1) continued development and characterization of large animal models for dystrophic cardiomyopathy. In light of recent success in creating rat, pig and monkey models using the CRISPR/Cas9 technology, model generation may no longer represent a formidable barrier as it was before214; (2) thorough evaluation of the most promising gene therapy strategies in large animal models215. Lack of solid large animal data has been an important factor limiting the translation of rodent study results to human patients. In this regard, there is an urgent need to thoroughly evaluate therapeutic efficacy in large mammals. For example, treating heart disease with tcDNA exon skipping and AAV micro-dystrophin gene therapy in dystrophin-deficient dogs216; (3) establishment of cardiac specific biomarkers that can be used to monitor disease progression and responses to gene therapy in animal models of dystrophic cardiomyopathy; (4) investigations of gene therapy for cardiac manifestations in muscular dystrophies other than DMD and LGMD2F. For many of these muscular dystrophies, gene therapy strategies have been developed for treating skeletal muscle myopathy. We need to test if similar approaches can attenuate cardiac disease.

In summary, gene therapy for dystrophic cardiomyopathy has taken a slow but steady path towards preclinical and eventually clinical studies. These efforts will undoubtedly be complicated by issues related to vector manufacturing, host immune responses, and the lack of enough patients for large-scale clinical trials due to the relatively low incidence of the disease. Nevertheless, we already have a solid foundation. The future of dystrophic cardiomyopathy gene therapy is very bright.

Highlights box.

  • Cardiomyopathy is a common complication in inherited muscular dystrophies.

  • Gene therapy holds great promise to reduce heart-related morbidity and mortality in muscular dystrophies.

  • Adeno-associated virus (AAV) is the most effective cardiac gene delivery vector.

  • Micro-dystrophin and sarcoglycan gene therapies have significantly improved the cardiac outcome in animal models of Duchenne muscular dystrophy and limb girdle muscular dystrophy, respectively.

  • Targeting pathogenic mechanisms with disease gene independent gene therapy opens exciting new opportunities.

  • Preclinical test in large animal models will pave the way to human trials.

Acknowledgments

D Duan is a member of the scientific advisory board for Solid GT, a subsidiary of Solid Biosciences. DMD research in the Duan lab is supported by the National Institutes of Health (NS-90634), Department of Defense (MD130014), Muscular Dystrophy Association, Parent Project Muscular Dystrophy, Jesse’s Journey-The Foundation for Gene and Cell Therapy, Hope for Javier and the University of Missouri. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Footnotes

Financial and competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

Contributor Information

Yongping Yue, Email: yuey@missouri.edu.

Ibrahim M. Binalsheikh, Email: binalsheikhi@health.missouri.edu.

Stacey B. Leach, Email: leachsb@missouri.edu.

Timothy L. Domeier, Email: domeiert@health.missouri.edu.

References

  • 1.Mercuri E, Muntoni F. Muscular dystrophies. Lancet. 2013 Mar 9;381(9869):845–60. doi: 10.1016/S0140-6736(12)61897-2. [DOI] [PubMed] [Google Scholar]
  • 2.Emery AE. The muscular dystrophies. Lancet. 2002 Feb 23;359(9307):687–95. doi: 10.1016/S0140-6736(02)07815-7. [DOI] [PubMed] [Google Scholar]
  • 3.Dellefave LM, McNally EM. Cardiomyopathy in neuromuscular disorders. Prog Ped Cardio. 2007;24:35–46. [Google Scholar]
  • 4.Hermans MC, Pinto YM, Merkies IS, de Die-Smulders CE, Crijns HJ, Faber CG. Hereditary muscular dystrophies and the heart. Neuromuscul Disord. 2010 Aug;20(8):479–92. doi: 10.1016/j.nmd.2010.04.008. [DOI] [PubMed] [Google Scholar]
  • 5.Cox GF, Kunkel LM. Dystrophies and heart disease. Current opinion in cardiology. 1997;12(3):329–43. [PubMed] [Google Scholar]
  • 6.Goodwin FC, Muntoni F. Cardiac involvement in muscular dystrophies: molecular mechanisms. Muscle Nerve. 2005 Nov;32(5):577–88. doi: 10.1002/mus.20352. [DOI] [PubMed] [Google Scholar]
  • 7.Finsterer J, Stollberger C. Cardiac involvement in primary myopathies. Cardiology. 2000;94(1):1–11. doi: 10.1159/000007039. [DOI] [PubMed] [Google Scholar]
  • 8.Silva MC, Meira ZM, Gurgel Giannetti J, da Silva MM, Campos AF, Barbosa Mde M, et al. Myocardial delayed enhancement by magnetic resonance imaging in patients with muscular dystrophy. J Am Coll Cardiol. 2007 May 8;49(18):1874–9. doi: 10.1016/j.jacc.2006.10.078. [DOI] [PubMed] [Google Scholar]
  • 9.Bilchick KC, Salerno M, Plitt D, Dori Y, Crawford TO, Drachman D, et al. Prevalence and distribution of regional scar in dysfunctional myocardial segments in Duchenne muscular dystrophy. Journal of cardiovascular magnetic resonance: official journal of the Society for Cardiovascular Magnetic Resonance. 2011;13:20. doi: 10.1186/1532-429X-13-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tandon A, Villa CR, Hor KN, Jefferies JL, Gao Z, Towbin JA, et al. Myocardial fibrosis burden predicts left ventricular ejection fraction and is associated with age and steroid treatment duration in Duchenne muscular dystrophy. Journal of the American Heart Association. 2015 Apr;4(4) doi: 10.1161/JAHA.114.001338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rahimov F, Kunkel LM. The cell biology of disease: cellular and molecular mechanisms underlying muscular dystrophy. J Cell Biol. 2013 May 13;201(4):499–510. doi: 10.1083/jcb.201212142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Guiraud S, Aartsma-Rus A, Vieira NM, Davies KE, van Ommen GJ, Kunkel LM. The pathogenesis and therapy of muscular dystrophies. Annual review of genomics and human genetics. 2015 Aug 24;16:281–308. doi: 10.1146/annurev-genom-090314-025003. [DOI] [PubMed] [Google Scholar]
  • 13.McNally EM, Kaltman JR, Benson DW, Canter CE, Cripe LH, Duan D, et al. Contemporary cardiac issues in Duchenne muscular dystrophy. Circulation. 2015 May 5;131(18):1590–8. doi: 10.1161/CIRCULATIONAHA.114.015151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wasala NB, Shin JH, Duan D. The evolution of heart gene delivery vectors. J Gene Med. 2011 Oct;13(10):557–65. doi: 10.1002/jgm.1600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Carter BJ. Adeno-associated virus and the development of adeno-associated virus vectors: a historical perspective. Mol Ther. 2004 Dec;10(6):981–9. doi: 10.1016/j.ymthe.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 16.Lai Y, Yue Y, Duan D. Evidence for the failure of adeno-associated virus serotype 5 to package a viral genome > or = 8.2 kb. Mol Ther. 2010 Jan;18(1):75–9. doi: 10.1038/mt.2009.256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ghosh A, Duan D. Expending adeno-associated viral vector capacity: a tale of two vectors. Biotechnology and Genetic Engineering Reviews. 2007;24:165–77. doi: 10.1080/02648725.2007.10648098. [DOI] [PubMed] [Google Scholar]
  • 18.Ghosh A, Yue Y, Lai Y, Duan D. A hybrid vector system expands aden-associated viral vector packaging capacity in a transgene independent manner. Mol Ther. 2008 Jan;16(1):124–30. doi: 10.1038/sj.mt.6300322. [DOI] [PubMed] [Google Scholar]
  • 19.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. Nature biotechnology. 2005 Nov;23(11):1435–9. doi: 10.1038/nbt1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Zhang Y, Yue Y, Li L, Hakim CH, Zhang K, Thomas GD, et al. Dual AAV therapy ameliorates exercise-induced muscle injury and functional ischemia in murine models of Duchenne muscular dystrophy. Hum Mol Genet. 2013 May 24;22(18):3720–9. doi: 10.1093/hmg/ddt224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Odom GL, Gregorevic P, Allen JM, Chamberlain JS. Gene therapy of mdx mice with large truncated dystrophins generated by recombination using rAAV6. Mol Ther. 2011 Sep 21;19(1):36–45. doi: 10.1038/mt.2010.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lostal W, Kodippili K, Yue Y, Duan D. Full-length dystrophin reconstitution with adeno-associated viral vectors. Hum Gene Ther. 2014 Mar 31;25(6):552–62. doi: 10.1089/hum.2013.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Toyo-oka T, Kawada T, Xi H, Nakazawa M, Masui F, Hemmi C, et al. Gene therapy prevents disruption of dystrophin-related proteins in a model of hereditary dilated cardiomyopathy in hamsters. Heart Lung Circ. 2002;11(3):174–81. doi: 10.1046/j.1444-2892.2002.00151.x. [DOI] [PubMed] [Google Scholar]
  • 24*.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 Sep 30;108(13):1626–32. doi: 10.1161/01.CIR.0000089371.11664.27. This is the first study demonstrating the potential of micro-dystrophin gene therapy to treat Duchenne cardiomyopathy in an animal model. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25*.Hoshijima M, Ikeda Y, Iwanaga Y, Minamisawa S, Date MO, Gu Y, et al. Chronic suppression of heart-failure progression by a pseudophosphorylated mutant of phospholamban via in vivo cardiac rAAV gene delivery. Nature medicine. 2002 Aug;8(8):864–71. doi: 10.1038/nm739. This study suggests that restoration of calcium homeostasis in the heart is a promising strategy to treat dystrophic cardiomyopathy. [DOI] [PubMed] [Google Scholar]
  • 26.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 Oct;10(21):1807–13. doi: 10.1038/sj.gt.3302078. [DOI] [PubMed] [Google Scholar]
  • 27.Kotterman MA, Schaffer DV. Engineering adeno-associated viruses for clinical gene therapy. Nature reviews Genetics. 2014 Jul;15(7):445–51. doi: 10.1038/nrg3742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28*.Gregorevic P, Blankinship MJ, Allen JM, Crawford RW, Meuse L, Miller DG, et al. Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nature medicine. 2004 Aug;10(8):828–34. doi: 10.1038/nm1085. This is the first study demonstrating systemic gene delivery in an animal model of muscular dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.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. Nature biotechnology. 2005 Mar;23(3):321–8. doi: 10.1038/nbt1073. [DOI] [PubMed] [Google Scholar]
  • 30.Pacak CA, Mah CS, Thattaliyath BD, Conlon TJ, Lewis MA, Cloutier DE, et al. Recombinant adeno-associated virus serotype 9 leads to preferential cardiac transduction in vivo. Circ Res. 2006 Aug 18;99(4):e3–9. doi: 10.1161/01.RES.0000237661.18885.f6. [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 Nov;14(22):1605–9. doi: 10.1038/sj.gt.3303029. [DOI] [PubMed] [Google Scholar]
  • 32.Nance ME, Duan D. Perspective on adeno-associated virus (AAV) capsid modification for Duchenne muscular dystrophy gene therapy. Hum Gene Ther. 2015 doi: 10.1089/hum.2015.107. In-press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.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 Jun;16(6):1073–80. doi: 10.1038/mt.2008.76. [DOI] [PubMed] [Google Scholar]
  • 34**.Bostick B, Shin J-H, Yue Y, Duan D. AAV-microdystrophin therapy improves cardiac performance in aged female mdx mice. Mol Ther. 2011;19(10):1826–32. doi: 10.1038/mt.2011.154. This is the first study demonstrating improvement of cardiac contractility following AAV-mediated micro-dystrophin gene therapy in a model of dilated Duchenne cardiomyopathy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35*.Bostick B, Shin JH, Yue Y, Wasala NB, Lai Y, Duan D. AAV micro-dystrophin gene therapy alleviates stress-induced cardiac death but not myocardial fibrosis in >21-m-old mdx mice, an end-stage model of Duchenne muscular dystrophy cardiomyopathy. J Mol Cell Cardiol. 2012 Aug;53(2):217–22. doi: 10.1016/j.yjmcc.2012.05.002. This study demonstrates that AAV micro-dystrophin gene therapy can reduce stress-induced cardiac death in terminal aged mdx mice. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bostick B, Yue Y, Lai Y, Long C, Li D, Duan D. Adeno-associated virus serotype-9 microdystrophin gene therapy ameliorates electrocardiographic abnormalities in mdx mice. Hum Gene Ther. 2008 Aug;19(8):851–6. doi: 10.1089/hum.2008.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ying Y, Muller OJ, Goehringer C, Leuchs B, Trepel M, Katus HA, et al. Heart-targeted adeno-associated viral vectors selected by in vivo biopanning of a random viral display peptide library. Gene Ther. 2010 Aug;17(8):980–90. doi: 10.1038/gt.2010.44. [DOI] [PubMed] [Google Scholar]
  • 38.Yang L, Jiang J, Drouin LM, Agbandje-McKenna M, Chen C, Qiao C, et al. A myocardium tropic adeno-associated virus (AAV) evolved by DNA shuffling and in vivo selection. Proc Natl Acad Sci U S A. 2009 Mar 10;106(10):3946–51. doi: 10.1073/pnas.0813207106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39*.Kunkel LM. 2004 William Allan award address. cloning of the DMD gene. Am J Hum Genet. 2005 Feb;76(2):205–14. doi: 10.1086/428143. This article provides an excellent review on the cloning of the dystrophin gene. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lai Y, Zhao J, Yue Y, Duan D. alpha2 and alpha3 helices of dystrophin R16 and R17 frame a microdomain in the alpha1 helix of dystrophin R17 for neuronal NOS binding. Proc Natl Acad Sci U S A. 2013 Nov 26;110(2):525–30. doi: 10.1073/pnas.1211431109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41*.Lai Y, Thomas GD, Yue Y, Yang HT, Li D, Long C, et al. Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy. J Clin Invest. 2009 Mar;119(3):624–35. doi: 10.1172/JCI36612. This study demonstraes the importance of dystrophin spectrin-like repeats 16 and 17 for Duchenne muscular dystrophy gene therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rybakova IN, Amann KJ, Ervasti JM. A new model for the interaction of dystrophin with F-actin. J Cell Biol. 1996;135(3):661–72. doi: 10.1083/jcb.135.3.661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Belanto JJ, Mader TL, Eckhoff MD, Strandjord DM, Banks GB, Gardner MK, et al. Microtubule binding distinguishes dystrophin from utrophin. Proc Natl Acad Sci U S A. 2014 Apr 15;111(15):5723–8. doi: 10.1073/pnas.1323842111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44*.England SB, Nicholson LV, Johnson MA, Forrest SM, Love DR, Zubrzycka-Gaarn EE, et al. Very mild muscular dystrophy associated with the deletion of 46% of dystrophin. Nature. 1990;343(6254):180–2. doi: 10.1038/343180a0. This study demonstrates for the first time that an abbreviated dystrophin gene may benefit DMD patients. [DOI] [PubMed] [Google Scholar]
  • 45.Phelps SF, Hauser MA, Cole NM, Rafael JA, Hinkle RT, Faulkner JA, et al. Expression of full-length and truncated dystrophin mini-genes in transgenic mdx mice. Hum Mol Genet. 1995;4(8):1251–8. doi: 10.1093/hmg/4.8.1251. [DOI] [PubMed] [Google Scholar]
  • 46.Wells DJ, Wells KE, Asante EA, Turner G, Sunada Y, Campbell KP, et al. Expression of human full-length and minidystrophin in transgenic mdx mice: implications for gene therapy of Duchenne muscular dystrophy. Hum Mol Genet. 1995;4(8):1245–50. doi: 10.1093/hmg/4.8.1245. [DOI] [PubMed] [Google Scholar]
  • 47**.Harper SQ, Hauser MA, DelloRusso C, Duan D, Crawford RW, Phelps SF, et al. Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy. Nature medicine. 2002;8(3):253–61. doi: 10.1038/nm0302-253. This study reveals the critical structure-funciton relationship of the dystrophin gene and the implication for Duchenne muscular dystrophy gene therapy. [DOI] [PubMed] [Google Scholar]
  • 48.Bostick B, Yue Y, Long C, Marschalk N, Fine DM, Chen J, et al. Cardiac expression of a mini-dystrophin that normalizes skeletal muscle force only partially restores heart function in aged mdx mice. Mol Ther. 2009 Feb;17(2):253–61. doi: 10.1038/mt.2008.264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Lai Y, Yue Y, Bostick B, Duan D. Delivering large therapeutic genes for muscle gene therapy. In: Duan D, editor. Muscle gene therapy. New York: Springer Science + Business Media, LLC; 2010. pp. 205–18. [Google Scholar]
  • 50.Duan D. From the smallest virus to the biggest gene: marching towards gene therapy for Duchenne muscular dystrophy. Discovery medicine. 2006 Jun;6(33):103–8. [PMC free article] [PubMed] [Google Scholar]
  • 51.Zhang Y, Duan D. Novel mini-dystrophin gene dual adeno-associated virus vectors restore neuronal nitric oxide synthase expression at the sarcolemma. Hum Gene Ther. 2012 Oct 24;23(1):98–103. doi: 10.1089/hum.2011.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Ghosh A, Yue Y, Duan D. Efficient transgene reconstitution with hybrid dual AAV vectors carrying the minimized bridging sequence. Hum Gene Ther. 2011;22(1):77–83. doi: 10.1089/hum.2010.122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ghosh A, Yue Y, Shin J-H, Duan D. Systemic trans-splicing AAV delivery efficiently transduces the heart of adult mdx mouse, a model for Duchenne muscular dystrophy. Hum Gene Ther. 2009;20(11):1319–28. doi: 10.1089/hum.2009.058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gregorevic P, Allen JM, Minami E, Blankinship MJ, Haraguchi M, Meuse L, et al. rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nature medicine. 2006 Jul;12(7):787–9. doi: 10.1038/nm1439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.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(25):13714–9. doi: 10.1073/pnas.240335297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Liu M, Yue Y, Harper SQ, Grange RW, Chamberlain JS, Duan D. Adeno-associated virus-mediated microdystrophin expression protects young mdx muscle from contraction-induced injury. Mol Ther. 2005 Feb;11(2):245–56. doi: 10.1016/j.ymthe.2004.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57**.Yue Y, Pan X, Hakim CH, Kodippili K, Zhang K, Shin JH, et al. Safe and bodywide muscle transduction in young adult Duchenne muscular dystrophy dogs with adeno-associated virus. Hum Mol Genet. 2015;24(20):5880–90. doi: 10.1093/hmg/ddv310. This study opens the door of systemic AAV gene therapy in a large mammal with muscular dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Shin JH, Pan X, Hakim CH, Yang HT, Yue Y, Zhang K, et al. Microdystrophin ameliorates muscular dystrophy in the canine model of Duchenne muscular dystrophy. Mol Ther. 2013 Jan 15;21(4):750–7. doi: 10.1038/mt.2012.283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Koo T, Okada T, Athanasopoulos T, Foster H, Takeda S, Dickson G. Long-term functional adeno-associated virus-microdystrophin expression in the dystrophic CXMDj dog. J Gene Med. 2011 Sep;13(9):497–506. doi: 10.1002/jgm.1602. [DOI] [PubMed] [Google Scholar]
  • 60.Townsend D, Blankinship MJ, Allen JM, Gregorevic P, Chamberlain JS, Metzger JM. Systemic administration of micro-dystrophin restores cardiac geometry and prevents dobutamine-induced cardiac pump failure. Mol Ther. 2007 Jun;15(6):1086–92. doi: 10.1038/sj.mt.6300144. [DOI] [PubMed] [Google Scholar]
  • 61.Schinkel S, Bauer R, Bekeredjian R, Stucka R, Rutschow D, Lochmuller H, et al. Long-term preservation of cardiac structure and function after adeno-associated virus serotype 9-mediated microdystrophin gene transfer in mdx mice. Hum Gene Ther. 2012 Jun;23(6):566–75. doi: 10.1089/hum.2011.017. [DOI] [PubMed] [Google Scholar]
  • 62.Shin JH, Nitahara-Kasahara Y, Hayashita-Kinoh H, Ohshima-Hosoyama S, Kinoshita K, Chiyo T, et al. Improvement of cardiac fibrosis in dystrophic mice by rAAV9-mediated microdystrophin transduction. Gene Ther. 2011 Sep;18(9):910–9. doi: 10.1038/gt.2011.36. [DOI] [PubMed] [Google Scholar]
  • 63*.Bostick B, Yue Y, Long C, Duan D. Prevention of dystrophin-deficient cardiomyopathy in twenty-one-month-old carrier mice by mosaic dystrophin expression or complementary dystrophin/utrophin expression. Circ Res. 2008 Jan 4;102(1):121–30. doi: 10.1161/CIRCRESAHA.107.162982. This study demonstrates that genetic correction of 50% cardiomyocytes may be sufficient to treat dystrophic cardiomyopathy. This study also demonstrates that aged female mdx mice have dialted cardiomyopathy. [DOI] [PubMed] [Google Scholar]
  • 64.Bostick B, Yue Y, Duan D. Gender influences cardiac function in the mdx model of Duchenne cardiomyopathy. Muscle Nerve. 2010 Oct;42(4):600–3. doi: 10.1002/mus.21763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Chamberlain JS, Metzger J, Reyes M, Townsend D, Faulkner JA. Dystrophin-deficient mdx mice display a reduced life span and are susceptible to spontaneous rhabdomyosarcoma. Faseb J. 2007 Jul;21(9):2195–204. doi: 10.1096/fj.06-7353com. [DOI] [PubMed] [Google Scholar]
  • 66.Li D, Long C, Yue Y, Duan D. Sub-physiological sarcoglycan expression contributes to compensatory muscle protection in mdx mice. Hum Mol Genet. 2009 Apr 1;18(7):1209–20. doi: 10.1093/hmg/ddp015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Aartsma-Rus A. Overview on DMD exon skipping. Methods Mol Biol. 2012;867:97–116. doi: 10.1007/978-1-61779-767-5_7. [DOI] [PubMed] [Google Scholar]
  • 68.Goemans NM, Tulinius M, van den Akker JT, Burm BE, Ekhart PF, Heuvelmans N, et al. Systemic Administration of PRO051 in Duchenne’s Muscular Dystrophy. N Engl J Med. 2011 Mar 23;364(16):1513–22. doi: 10.1056/NEJMoa1011367. [DOI] [PubMed] [Google Scholar]
  • 69*.van Deutekom JC, Janson AA, Ginjaar IB, Frankhuizen WS, Aartsma-Rus A, Bremmer-Bout M, et al. Local dystrophin restoration with antisense oligonucleotide PRO051. N Engl J Med. 2007 Dec 27;357(26):2677–86. doi: 10.1056/NEJMoa073108. This is the first study demonstrating exon-skipping in human patients. [DOI] [PubMed] [Google Scholar]
  • 70.Cirak S, Arechavala-Gomeza V, Guglieri M, Feng L, Torelli S, Anthony K, et al. Exon skipping and dystrophin restoration in patients with Duchenne muscular dystrophy after systemic phosphorodiamidate morpholino oligomer treatment: an open-label, phase 2, dose-escalation study. Lancet. 2011 Aug 13;378(9791):595–605. doi: 10.1016/S0140-6736(11)60756-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kinali M, Arechavala-Gomeza V, Feng L, Cirak S, Hunt D, Adkin C, et al. Local restoration of dystrophin expression with the morpholino oligomer AVI-4658 in Duchenne muscular dystrophy: a single-blind, placebo-controlled, dose-escalation, proof-of-concept study. Lancet Neurol. 2009 Oct;8(10):918–28. doi: 10.1016/S1474-4422(09)70211-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mendell J, Rodino-Klapac LR, Sahenk Z, Roush K, Bird L, Lowes LP, et al. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013 Aug 1;74(5):637–47. doi: 10.1002/ana.23982. [DOI] [PubMed] [Google Scholar]
  • 73.Voit T, Topaloglu H, Straub V, Muntoni F, Deconinck N, Campion G, et al. Safety and efficacy of drisapersen for the treatment of Duchenne muscular dystrophy (DEMAND II): an exploratory, randomised, placebo-controlled phase 2 study. Lancet Neurol. 2014 Oct;13(10):987–96. doi: 10.1016/S1474-4422(14)70195-4. [DOI] [PubMed] [Google Scholar]
  • 74.Alter J, Lou F, Rabinowitz A, Yin H, Rosenfeld J, Wilton SD, et al. Systemic delivery of morpholino oligonucleotide restores dystrophin expression bodywide and improves dystrophic pathology. Nature medicine. 2006 Feb;12(2):175–7. doi: 10.1038/nm1345. [DOI] [PubMed] [Google Scholar]
  • 75.Malerba A, Boldrin L, Dickson G. Long-term systemic administration of unconjugated morpholino oligomers for therapeutic expression of dystrophin by exon skipping in skeletal muscle: implications for cardiac muscle integrity. Nucleic Acid Ther. 2011 Aug;21(4):293–8. doi: 10.1089/nat.2011.0306. [DOI] [PubMed] [Google Scholar]
  • 76.Crisp A, Yin H, Goyenvalle A, Betts C, Moulton HM, Seow Y, et al. Diaphragm rescue alone prevents heart dysfunction in dystrophic mice. Hum Mol Genet. 2011 Feb 1;20(3):413–21. doi: 10.1093/hmg/ddq477. [DOI] [PubMed] [Google Scholar]
  • 77.Wu B, Moulton HM, Iversen PL, Jiang J, Li J, Spurney CF, et al. Effective rescue of dystrophin improves cardiac function in dystrophin-deficient mice by a modified morpholino oligomer. Proc Natl Acad Sci U S A. 2008 Sep 30;105(39):14814–9. doi: 10.1073/pnas.0805676105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Yin H, Lu Q, Wood M. Effective exon skipping and restoration of dystrophin expression by peptide nucleic acid antisense oligonucleotides in mdx mice. Mol Ther. 2008 Jan;16(1):38–45. doi: 10.1038/sj.mt.6300329. [DOI] [PubMed] [Google Scholar]
  • 79.Yin H, Moulton HM, Seow Y, Boyd C, Boutilier J, Iverson P, et al. Cell-penetrating peptide-conjugated antisense oligonucleotides restore systemic muscle and cardiac dystrophin expression and function. Hum Mol Genet. 2008 Dec 15;17(24):3909–18. doi: 10.1093/hmg/ddn293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Jearawiriyapaisarn N, Moulton HM, Buckley B, Roberts J, Sazani P, Fucharoen S, et al. Sustained dystrophin expression induced by peptide-conjugated morpholino oligomers in the muscles of mdx mice. Mol Ther. 2008 Sep;16(9):1624–9. doi: 10.1038/mt.2008.120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Wu B, Li Y, Morcos PA, Doran TJ, Lu P, Lu QL. Octa-guanidine morpholino restores dystrophin expression in cardiac and skeletal muscles and ameliorates pathology in dystrophic mdx mice. Mol Ther. 2009 Mar 10;17(5):864–71. doi: 10.1038/mt.2009.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Ferlini A, Sabatelli P, Fabris M, Bassi E, Falzarano S, Vattemi G, et al. Dystrophin restoration in skeletal, heart and skin arrector pili smooth muscle of mdx mice by ZM2 NP-AON complexes. Gene Ther. 2010 Mar;17(3):432–8. doi: 10.1038/gt.2009.145. [DOI] [PubMed] [Google Scholar]
  • 83.Jearawiriyapaisarn N, Moulton HM, Sazani P, Kole R, Willis MS. Long-term improvement in mdx cardiomyopathy after therapy with peptide-conjugated morpholino oligomers. Cardiovasc Res. 2010 Feb 1;85(3):444–53. doi: 10.1093/cvr/cvp335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yin H, Moulton HM, Betts C, Seow Y, Boutilier J, Iverson PL, et al. A fusion peptide directs enhanced systemic dystrophin exon skipping and functional restoration in dystrophin-deficient mdx mice. Hum Mol Genet. 2009 Nov 15;18(22):4405–14. doi: 10.1093/hmg/ddp395. [DOI] [PubMed] [Google Scholar]
  • 85.Yin H, Moulton HM, Betts C, Merritt T, Seow Y, Ashraf S, et al. Functional rescue of dystrophin-deficient mdx mice by a chimeric peptide-PMO. Mol Ther. 2010 Oct;18(10):1822–9. doi: 10.1038/mt.2010.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Yin H, Saleh AF, Betts C, Camelliti P, Seow Y, Ashraf S, et al. Pip5 transduction peptides direct high efficiency oligonucleotide-mediated dystrophin exon skipping in heart and phenotypic correction in mdx mice. Mol Ther. 2011 Jul;19(7):1295–303. doi: 10.1038/mt.2011.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wu B, Xiao B, Cloer C, Shaban M, Sali A, Lu P, et al. One-year treatment of morpholino antisense oligomer improves skeletal and cardiac muscle functions in dystrophic mdx mice. Mol Ther. 2011 Mar;19(3):576–83. doi: 10.1038/mt.2010.288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Amantana A, Moulton HM, Cate ML, Reddy MT, Whitehead T, Hassinger JN, et al. Pharmacokinetics, biodistribution, stability and toxicity of a cell-penetrating peptide-morpholino oligomer conjugate. Bioconjug Chem. 2007 Jul-Aug;18(4):1325–31. doi: 10.1021/bc070060v. [DOI] [PubMed] [Google Scholar]
  • 89*.Goyenvalle A, Griffith G, Babbs A, El Andaloussi S, Ezzat K, Avril A, et al. Functional correction in mouse models of muscular dystrophy using exon-skipping tricyclo-DNA oligomers. Nature medicine. 2015 Mar;21(3):270–5. doi: 10.1038/nm.3765. This study demonstrates that newly developed tricyclo oligomers can significantly improve exon-skipping in animal models of muscular dystrophy. [DOI] [PubMed] [Google Scholar]
  • 90.Goyenvalle A, Babbs A, Wright J, Wilkins V, Powell D, Garcia L, et al. Rescue of severely affected dystrophin/utrophin-deficient mice through scAAV-U7snRNA-mediated exon skipping. Hum Mol Genet. 2012 Jun 1;21(11):2559–71. doi: 10.1093/hmg/dds082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91*.Bish LT, Sleeper MM, Forbes SC, Wang B, Reynolds C, Singletary GE, et al. Long-term restoration of cardiac dystrophin expression in golden retriever muscular dystrophy following rAAV6-mediated exon skipping. Mol Ther. 2012 Mar;20(3):580–9. doi: 10.1038/mt.2011.264. This study suggests that AAV-mediated exon-skipping can treat dystrophic cardiomyopathy in a large animal model. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Barbash IM, Cecchini S, Faranesh AZ, Virag T, Li L, Yang Y, et al. MRI roadmap-guided transendocardial delivery of exon-skipping recombinant adeno-associated virus restores dystrophin expression in a canine model of Duchenne muscular dystrophy. Gene Ther. 2013 Mar;20(3):274–82. doi: 10.1038/gt.2012.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gaj T, Gersbach CA, Barbas CF., 3rd ZFN, TALEN, and CRISPR/Cas-based methods for genome engineering. Trends in biotechnology. 2013 Jul;31(7):397–405. doi: 10.1016/j.tibtech.2013.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94*.Ousterout DG, Kabadi AM, Thakore PI, Majoros WH, Reddy TE, Gersbach CA. Multiplex CRISPR/Cas9-based genome editing for correction of dystrophin mutations that cause Duchenne muscular dystrophy. Nat Commun. 2015;6:6244. doi: 10.1038/ncomms7244. This study demonstrates the feasiblity of genome editing as a therapy for Duchenne muscular dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ousterout DG, Kabadi AM, Thakore PI, Perez-Pinera P, Brown MT, Majoros WH, et al. Correction of dystrophin expression in cells from Duchenne muscular dystrophy patients through genomic excision of exon 51 by zinc finger nucleases. Mol Ther. 2014 Dec 10; doi: 10.1038/mt.2014.234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ousterout DG, Perez-Pinera P, Thakore PI, Kabadi AM, Brown MT, Qin X, et al. Reading frame correction by targeted genome editing restores dystrophin expression in cells from Duchenne muscular dystrophy patients. Mol Ther. 2013 Sep;21(9):1718–26. doi: 10.1038/mt.2013.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Nelson CE, Ousterout DG, Robinson-Hamm J, A ME, Castellanos RM, Ran FA, et al. Correction of the dystrophin gene by the CRISPR/Cas9 system in a mouse model of muscular dystrophy. Mol Ther. 2015;23(Supp 1):S157–8. [Google Scholar]
  • 98.Nigro V, Savarese M. Genetic basis of limb-girdle muscular dystrophies: the 2014 update. Acta myologica: myopathies and cardiomyopathies: official journal of the Mediterranean Society of Myology/edited by the Gaetano Conte Academy for the study of striated muscle diseases. 2014 May;33(1):1–12. [PMC free article] [PubMed] [Google Scholar]
  • 99.Guglieri M, Straub V, Bushby K, Lochmuller H. Limb-girdle muscular dystrophies. Current opinion in neurology. 2008 Oct;21(5):576–84. doi: 10.1097/WCO.0b013e32830efdc2. [DOI] [PubMed] [Google Scholar]
  • 100.Wallace LM, Garwick SE, Harper SQ. RNAi therapy for dominant muscular dystrophies and other myopathies. In: Duan D, editor. Muscle gene therapy. New York: Springer Science + Business Media, LLC; 2010. pp. 99–115. [Google Scholar]
  • 101.Liu J, Harper SQ. RNAi-based gene therapy for dominant limb girdle muscular dystrophies. Current gene therapy. 2012 Aug;12(4):307–14. doi: 10.2174/156652312802083585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102*.Liu J, Wallace LM, Garwick-Coppens SE, Sloboda DD, Davis CS, Hakim CH, et al. RNAi-mediated gene silencing of mutant myotilin improves myopathy in LGMD1A mice. Molecular therapy Nucleic acids. 2014;3:e160. doi: 10.1038/mtna.2014.13. This study reveals the potential of using RNA interference to treat dominant muscular dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Han R, Bansal D, Miyake K, Muniz VP, Weiss RM, McNeil PL, et al. Dysferlin-mediated membrane repair protects the heart from stress-induced left ventricular injury. J Clin Invest. 2007 Jul;117(7):1805–13. doi: 10.1172/JCI30848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Chase TH, Cox GA, Burzenski L, Foreman O, Shultz LD. Dysferlin deficiency and the development of cardiomyopathy in a mouse model of limb-girdle muscular dystrophy 2B. Am J Pathol. 2009 Dec;175(6):2299–308. doi: 10.2353/ajpath.2009.080930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wenzel K, Geier C, Qadri F, Hubner N, Schulz H, Erdmann B, et al. Dysfunction of dysferlin-deficient hearts. Journal of molecular medicine. 2007 Nov;85(11):1203–14. doi: 10.1007/s00109-007-0253-7. [DOI] [PubMed] [Google Scholar]
  • 106.Kuru S, Yasuma F, Wakayama T, Kimura S, Konagaya M, Aoki M, et al. A patient with limb girdle muscular dystrophy type 2B (LGMD2B) manifesting cardiomyopathy. Rinsho shinkeigaku = Clinical neurology. 2004 Jun;44(6):375–8. [PubMed] [Google Scholar]
  • 107.Lostal W, Bartoli M, Bourg N, Roudaut C, Bentaib A, Miyake K, et al. Efficient recovery of dysferlin deficiency by dual adeno-associated vector-mediated gene transfer. Hum Mol Genet. 2010 May 15;19(10):1897–907. doi: 10.1093/hmg/ddq065. [DOI] [PubMed] [Google Scholar]
  • 108.Krahn M, Wein N, Bartoli M, Lostal W, Courrier S, Bourg-Alibert N, et al. A naturally occurring human minidysferlin protein repairs sarcolemmal lesions in a mouse model of dysferlinopathy. Sci Transl Med. 2010 Sep 22;2(50):50ra69. doi: 10.1126/scitranslmed.3000951. [DOI] [PubMed] [Google Scholar]
  • 109.Aartsma-Rus A, Singh KH, Fokkema IF, Ginjaar IB, van Ommen GJ, den Dunnen JT, et al. Therapeutic exon skipping for dysferlinopathies? European journal of human genetics: EJHG. 2010 Aug;18(8):889–94. doi: 10.1038/ejhg.2010.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Pryadkina M, Lostal W, Bourg N, Charton K, Roudaut C, Hirsch ML, et al. A comparison of AAV strategies distinguishes overlapping vectors for efficient systemic delivery of the 6.2 kb Dysferlin coding sequence. Molecular therapy Methods & clinical development. 2015;2:15009. doi: 10.1038/mtm.2015.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Philippi S, Lorain S, Beley C, Peccate C, Precigout G, Spuler S, et al. Dysferlin rescue by spliceosome-mediated pre-mRNA trans-splicing targeting introns harbouring weakly defined 3′ splice sites. Hum Mol Genet. 2015 Jul 15;24(14):4049–60. doi: 10.1093/hmg/ddv141. [DOI] [PubMed] [Google Scholar]
  • 112.Sondergaard PC, Griffin DA, Pozsgai ER, Johnson RW, Grose WE, Heller KN, et al. AAV.Dysferlin overlap vectors restore function in dysferlinopathy animal models. Annals of clinical and translational neurology. 2015 Mar;2(3):256–70. doi: 10.1002/acn3.172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Wein N, Avril A, Bartoli M, Beley C, Chaouch S, Laforet P, et al. Efficient bypass of mutations in dysferlin deficient patient cells by antisense-induced exon skipping. Hum Mutat. 2010 Feb;31(2):136–42. doi: 10.1002/humu.21160. [DOI] [PubMed] [Google Scholar]
  • 114.Lostal W, Bartoli M, Roudaut C, Bourg N, Krahn M, Pryadkina M, et al. Lack of correlation between outcomes of membrane repair assay and correction of dystrophic changes in experimental therapeutic strategy in dysferlinopathy. PLoS One. 2012;7(5):e38036. doi: 10.1371/journal.pone.0038036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Stedman H, Wilson JM, Finke R, Kleckner AL, Mendell J. Phase I clinical trial utilizing gene therapy for limb girdle muscular dystrophy: alpha-, beta-, gamma-, or delta-sarcoglycan gene delivered with intramuscular instillations of adeno-associated vectors. Hum Gene Ther. 2000;11(5):777–90. doi: 10.1089/10430340050015671. [DOI] [PubMed] [Google Scholar]
  • 116.Herson S, Hentati F, Rigolet A, Behin A, Romero NB, Leturcq F, et al. A phase I trial of adeno-associated virus serotype 1-gamma-sarcoglycan gene therapy for limb girdle muscular dystrophy type 2C. Brain. 2012 Feb;135(Pt 2):483–92. doi: 10.1093/brain/awr342. [DOI] [PubMed] [Google Scholar]
  • 117.Mendell JR, Rodino-Klapac LR, Rosales XQ, Coley BD, Galloway G, Lewis S, et al. Sustained alpha-sarcoglycan gene expression after gene transfer in limb-girdle muscular dystrophy, type 2D. Ann Neurol. 2010 Nov;68(5):629–38. doi: 10.1002/ana.22251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118*.Mendell JR, Rodino-Klapac LR, Rosales-Quintero X, Kota J, Coley BD, Galloway G, et al. Limb-girdle muscular dystrophy type 2D gene therapy restores alpha-sarcoglycan and associated proteins. Ann Neurol. 2009 Sep;66(3):290–7. doi: 10.1002/ana.21732. This study demonstrates the therapeutic potential of AAV therapy for limb girdle muscular dystrophy in human patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Dressman D, Araishi K, Imamura M, Sasaoka T, Liu LA, Engvall E, et al. Delivery of alpha- and beta-sarcoglycan by recombinant adeno-associated virus: efficient rescue of muscle, but differential toxicity. Hum Gene Ther. 2002;13(13):1631–46. doi: 10.1089/10430340260201725. [DOI] [PubMed] [Google Scholar]
  • 120.Pozsgai ER, Griffin DA, Heller KN, Mendell JR, Rodino-Klapac LR. beta-Sarcoglycan gene transfer decreases fibrosis and restores force in LGMD2E mice. Gene Ther. 2015 Aug 20; doi: 10.1038/gt.2015.80. [DOI] [PubMed] [Google Scholar]
  • 121*.Zhu T, Zhou L, Mori S, Wang Z, McTiernan CF, Qiao C, et al. Sustained whole-body functional rescue in congestive heart failure and muscular dystrophy hamsters by systemic gene transfer. Circulation. 2005 Oct 25;112(17):2650–9. doi: 10.1161/CIRCULATIONAHA.105.565598. This study demonstrates the feasibility of systemic AAV gene therapy to treat heart disease associated with limb girdle muscular dystrophy. [DOI] [PubMed] [Google Scholar]
  • 122.Vitiello C, Faraso S, Sorrentino NC, Di Salvo G, Nusco E, Nigro G, et al. Disease rescue and increased lifespan in a model of cardiomyopathy and muscular dystrophy by combined AAV treatments. PLoS One. 2009;4(3):e5051. doi: 10.1371/journal.pone.0005051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Goehringer C, Rutschow D, Bauer R, Schinkel S, Weichenhan D, Bekeredjian R, et al. Prevention of cardiomyopathy in delta-sarcoglycan knockout mice after systemic transfer of targeted adeno-associated viral vectors. Cardiovasc Res. 2009 Jun 1;82(3):404–10. doi: 10.1093/cvr/cvp061. [DOI] [PubMed] [Google Scholar]
  • 124.Kawada T, Nakazawa M, Nakauchi S, Yamazaki K, Shimamoto R, Urabe M, et al. Rescue of hereditary form of dilated cardiomyopathy by rAAV-mediated somatic gene therapy: amelioration of morphological findings, sarcolemmal permeability, cardiac performances, and the prognosis of TO- 2 hamsters. Proc Natl Acad Sci U S A. 2002;99(2):901–6. doi: 10.1073/pnas.022641799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Hoshijima M, Hayashi T, Jeon YE, Fu Z, Gu Y, Dalton ND, et al. Delta-sarcoglycan gene therapy halts progression of cardiac dysfunction, improves respiratory failure, and prolongs life in myopathic hamsters. Circ Heart Fail. 2011 Jan;4(1):89–97. doi: 10.1161/CIRCHEARTFAILURE.110.957258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Poppe M, Bourke J, Eagle M, Frosk P, Wrogemann K, Greenberg C, et al. Cardiac and respiratory failure in limb-girdle muscular dystrophy 2I. Ann Neurol. 2004 Nov;56(5):738–41. doi: 10.1002/ana.20283. [DOI] [PubMed] [Google Scholar]
  • 127.Chan YM, Keramaris-Vrantsis E, Lidov HG, Norton JH, Zinchenko N, Gruber HE, et al. Fukutin-related protein is essential for mouse muscle, brain and eye development and mutation recapitulates the wide clinical spectrums of dystroglycanopathies. Hum Mol Genet. 2010 Oct 15;19(20):3995–4006. doi: 10.1093/hmg/ddq314. [DOI] [PubMed] [Google Scholar]
  • 128.Qiao C, Wang CH, Zhao C, Lu P, Awano H, Xiao B, et al. Muscle and heart function restoration in a limb girdle muscular dystrophy 2I (LGMD2I) mouse model by systemic FKRP gene delivery. Mol Ther. 2014 Nov;22(11):1890–9. doi: 10.1038/mt.2014.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Barresi R, Campbell KP. Dystroglycan: from biosynthesis to pathogenesis of human disease. J Cell Sci. 2006 Jan 15;119(Pt 2):199–207. doi: 10.1242/jcs.02814. [DOI] [PubMed] [Google Scholar]
  • 130.Godfrey C, Foley AR, Clement E, Muntoni F. Dystroglycanopathies: coming into focus. Curr Opin Genet Dev. 2011 Jun;21(3):278–85. doi: 10.1016/j.gde.2011.02.001. [DOI] [PubMed] [Google Scholar]
  • 131.Nakanishi T, Sakauchi M, Kaneda Y, Tomimatsu H, Saito K, Nakazawa M, et al. Cardiac involvement in Fukuyama-type congenital muscular dystrophy. Pediatrics. 2006 Jun;117(6):e1187–92. doi: 10.1542/peds.2005-2469. [DOI] [PubMed] [Google Scholar]
  • 132.Taniguchi-Ikeda M, Kobayashi K, Kanagawa M, Yu CC, Mori K, Oda T, et al. Pathogenic exon-trapping by SVA retrotransposon and rescue in Fukuyama muscular dystrophy. Nature. 2011 Oct 6;478(7367):127–31. doi: 10.1038/nature10456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Pane M, Messina S, Vasco G, Foley AR, Morandi L, Pegoraro E, et al. Respiratory and cardiac function in congenital muscular dystrophies with alpha dystroglycan deficiency. Neuromuscul Disord. 2012 Aug;22(8):685–9. doi: 10.1016/j.nmd.2012.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Xu L, Lu PJ, Wang CH, Keramaris E, Qiao C, Xiao B, et al. Adeno-associated virus 9 mediated FKRP gene therapy restores functional glycosylation of alpha-dystroglycan and improves muscle functions. Mol Ther. 2013 Oct;21(10):1832–40. doi: 10.1038/mt.2013.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Chaudhry SP, Frishman WH. Myotonic dystrophies and the heart. Cardiology in review. 2012 Jan-Feb;20(1):1–3. doi: 10.1097/CRD.0b013e31821950f9. [DOI] [PubMed] [Google Scholar]
  • 136.Gomes-Pereira M, Cooper TA, Gourdon G. Myotonic dystrophy mouse models: towards rational therapy development. Trends in molecular medicine. 2011 Sep;17(9):506–17. doi: 10.1016/j.molmed.2011.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137*.Mahadevan MS, Yadava RS, Yu Q, Balijepalli S, Frenzel-McCardell CD, Bourne TD, et al. Reversible model of RNA toxicity and cardiac conduction defects in myotonic dystrophy. Nature genetics. 2006 Sep;38(9):1066–70. doi: 10.1038/ng1857. This study shows the proof-of-principle for reversing cardiac defects in myotonic dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Orengo JP, Chambon P, Metzger D, Mosier DR, Snipes GJ, Cooper TA. Expanded CTG repeats within the DMPK 3′ UTR causes severe skeletal muscle wasting in an inducible mouse model for myotonic dystrophy. Proc Natl Acad Sci U S A. 2008 Feb 19;105(7):2646–51. doi: 10.1073/pnas.0708519105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Klein AF, Dastidar S, Furling D, Chuah MK. Therapeutic approaches for dominant muscle diseases: highlight on myotonic dystrophy. Current gene therapy. 2015;15(4):329–37. doi: 10.2174/1566523215666150630120537. [DOI] [PubMed] [Google Scholar]
  • 140.Gao Z, Cooper TA. Antisense oligonucleotides: rising stars in eliminating RNA toxicity in myotonic dystrophy. Hum Gene Ther. 2013 May;24(5):499–507. doi: 10.1089/hum.2012.212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Zhang W, Wang Y, Dong S, Choudhury R, Jin Y, Wang Z. Treatment of type 1 myotonic dystrophy by engineering site-specific RNA endonucleases that target (CUG)(n) repeats. Mol Ther. 2014 Feb;22(2):312–20. doi: 10.1038/mt.2013.251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Wheeler TM, Leger AJ, Pandey SK, MacLeod AR, Nakamori M, Cheng SH, et al. Targeting nuclear RNA for in vivo correction of myotonic dystrophy. Nature. 2012 Aug 2;488(7409):111–5. doi: 10.1038/nature11362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lee JE, Bennett CF, Cooper TA. RNase H-mediated degradation of toxic RNA in myotonic dystrophy type 1. Proc Natl Acad Sci U S A. 2012 Mar 13;109(11):4221–6. doi: 10.1073/pnas.1117019109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Wheeler TM, Sobczak K, Lueck JD, Osborne RJ, Lin X, Dirksen RT, et al. Reversal of RNA dominance by displacement of protein sequestered on triplet repeat RNA. Science. 2009 Jul 17;325(5938):336–9. doi: 10.1126/science.1173110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Blake DJ, Tinsley JM, Davies KE. Utrophin: a structural and functional comparison to dystrophin. Brain pathology. 1996;6(1):37–47. doi: 10.1111/j.1750-3639.1996.tb00781.x. [DOI] [PubMed] [Google Scholar]
  • 146.Li D, Bareja A, Judge L, Yue Y, Lai Y, Fairclough R, et al. Sarcolemmal nNOS anchoring reveals a qualitative difference between dystrophin and utrophin. J Cell Sci. 2010 Jun 15;123(Pt 12):2008–13. doi: 10.1242/jcs.064808. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Rybakova IN, Humston JL, Sonnemann KJ, Ervasti JM. Dystrophin and utrophin bind actin through distinct modes of contact. J Biol Chem. 2006 Apr 14;281(15):9996–10001. doi: 10.1074/jbc.M513121200. [DOI] [PubMed] [Google Scholar]
  • 148.Odom GL, Gregorevic P, Allen JM, Finn E, Chamberlain JS. Microutrophin delivery through rAAV6 increases lifespan and improves muscle function in dystrophic dystrophin/utrophin-deficient mice. Mol Ther. 2008 Sep;16(9):1539–45. doi: 10.1038/mt.2008.149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Strimpakos G, Corbi N, Pisani C, Di Certo MG, Onori A, Luvisetto S, et al. Novel adeno-associated viral vector delivering the utrophin gene regulator jazz counteracts dystrophic pathology in mdx mice. Journal of cellular physiology. 2014 Sep;229(9):1283–91. doi: 10.1002/jcp.24567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Peter AK, Marshall JL, Crosbie RH. Sarcospan reduces dystrophic pathology: stabilization of the utrophin-glycoprotein complex. J Cell Biol. 2008 Nov 3;183(3):419–27. doi: 10.1083/jcb.200808027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol. 2001;155(1):123–32. doi: 10.1083/jcb.200105110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152*.Lai Y, Zhao J, Yue Y, Wasala NB, Duan D. Partial restoration of cardiac function with ΔPDZ nNOS in aged mdx model of Duchenne cardiomyopathy. Hum Mol Genet. 2014 Feb 11;23(12):3189–99. doi: 10.1093/hmg/ddu029. This study demonstrates the feasibility of nNOS-based gene therapy for treating Duchenne cardiomyopathy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Burkin DJ, Kaufman SJ. The alpha7beta1 integrin in muscle development and disease. Cell and tissue research. 1999 Apr;296(1):183–90. doi: 10.1007/s004410051279. [DOI] [PubMed] [Google Scholar]
  • 154.Heller KN, Montgomery CL, Janssen PM, Clark KR, Mendell JR, Rodino-Klapac LR. AAV-mediated overexpression of human alpha7 integrin leads to histological and functional improvement in dystrophic mice. Mol Ther. 2013 Mar;21(3):520–5. doi: 10.1038/mt.2012.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Heller KN, Montgomery CL, Shontz KM, Clark KR, Mendell JR, Rodino-Klapac LR. Human alpha7 integrin gene (ITGA7) delivered by adeno-associated virus extends survival of severely affected dystrophin/utrophin-deficient mice. Hum Gene Ther. 2015 Aug 11; doi: 10.1089/hum.2015.062. online publication 08-11-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Burr AR, Molkentin JD. Genetic evidence in the mouse solidifies the calcium hypothesis of myofiber death in muscular dystrophy. Cell death and differentiation. 2015 Jun 19;22(9):1402–12. doi: 10.1038/cdd.2015.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Shin JH, Bostick B, Yue Y, Hajjar R, Duan D. SERCA2a gene transfer improves electrocardiographic performance in aged mdx mice. J Transl Med. 2011;9:132. doi: 10.1186/1479-5876-9-132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Wasala NB, Yue Y, Duan D. AAV-SERCA2a gene therapy ameliorated dystrophin phenotype in mdx mice. 2015 Association for Clinical and Translational Science (ACTS) Annual Conference; Washington, DC. 2015. [Google Scholar]
  • 159.Tang Y, Reay DP, Salay MN, Mi MY, Clemens PR, Guttridge DC, et al. Inhibition of the IKK/NF-kappaB pathway by AAV gene transfer improves muscle regeneration in older mdx mice. Gene Ther. 2010 Dec;17(12):1476–83. doi: 10.1038/gt.2010.110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Yang Q, Tang Y, Imbrogno K, Lu A, Proto JD, Chen A, et al. AAV-based shRNA silencing of NF-kappaB ameliorates muscle pathologies in mdx mice. Gene Ther. 2012 Dec;19(12):1196–204. doi: 10.1038/gt.2011.207. [DOI] [PubMed] [Google Scholar]
  • 161.Selsby JT, Morine KJ, Pendrak K, Barton ER, Sweeney HL. Rescue of dystrophic skeletal muscle by PGC-1alpha involves a fast to slow fiber type shift in the mdx mouse. PLoS One. 2012;7(1):e30063. doi: 10.1371/journal.pone.0030063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Xu R, Camboni M, Martin PT. Postnatal overexpression of the CT GalNAc transferase inhibits muscular dystrophy in mdx mice without altering muscle growth or neuromuscular development: evidence for a utrophin-independent mechanism. Neuromuscul Disord. 2007 Mar;17(3):209–20. doi: 10.1016/j.nmd.2006.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Bish LT, Sleeper MM, Forbes SC, Morine KJ, Reynolds C, Singletary GE, et al. Long-term systemic myostatin inhibition via liver-targeted gene transfer in golden retriever muscular dystrophy. Hum Gene Ther. 2011 Dec;22(12):1499–509. doi: 10.1089/hum.2011.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Haidet AM, Rizo L, Handy C, Umapathi P, Eagle A, Shilling C, et al. Long-term enhancement of skeletal muscle mass and strength by single gene administration of myostatin inhibitors. Proc Natl Acad Sci U S A. 2008 Mar 18;105(11):4318–22. doi: 10.1073/pnas.0709144105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Alexander MS, Casar JC, Motohashi N, Vieira NM, Eisenberg I, Marshall JL, et al. MicroRNA-486-dependent modulation of DOCK3/PTEN/AKT signaling pathways improves muscular dystrophy-associated symptoms. J Clin Invest. 2014 Jun 2;124(6):2651–67. doi: 10.1172/JCI73579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Bogdanovich S, Krag TO, Barton ER, Morris LD, Whittemore LA, Ahima RS, et al. Functional improvement of dystrophic muscle by myostatin blockade. Nature. 2002;420(6914):418–21. doi: 10.1038/nature01154. [DOI] [PubMed] [Google Scholar]
  • 167.Wagner KR, McPherron AC, Winik N, Lee SJ. Loss of myostatin attenuates severity of muscular dystrophy in mdx mice. Ann Neurol. 2002 Dec;52(6):832–6. doi: 10.1002/ana.10385. [DOI] [PubMed] [Google Scholar]
  • 168.Nakatani M, Takehara Y, Sugino H, Matsumoto M, Hashimoto O, Hasegawa Y, et al. Transgenic expression of a myostatin inhibitor derived from follistatin increases skeletal muscle mass and ameliorates dystrophic pathology in mdx mice. FASEB J. 2008 Feb;22(2):477–87. doi: 10.1096/fj.07-8673com. [DOI] [PubMed] [Google Scholar]
  • 169*.Mendell JR, Sahenk Z, Malik V, Gomez AM, Flanigan KM, Lowes LP, et al. A phase I/IIa follistatin gene therapy trial for Becker muscular dystrophy. Mol Ther. 2015 Oct 17;23(1):192–201. doi: 10.1038/mt.2014.200. This study demonstrates therapeutic potential of follistatin gene therapy in human patients. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Li ZF, Shelton GD, Engvall E. Elimination of myostatin does not combat muscular dystrophy in dy mice but increases postnatal lethality. Am J Pathol. 2005 Feb;166(2):491–7. doi: 10.1016/S0002-9440(10)62271-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Bogdanovich S, McNally EM, Khurana TS. Myostatin blockade improves function but not histopathology in a murine model of limb-girdle muscular dystrophy 2C. Muscle Nerve. 2008 Mar;37(3):308–16. doi: 10.1002/mus.20920. [DOI] [PubMed] [Google Scholar]
  • 172.Parsons SA, Millay DP, Sargent MA, McNally EM, Molkentin JD. Age-dependent effect of myostatin blockade on disease severity in a murine model of limb-girdle muscular dystrophy. Am J Pathol. 2006 Jun;168(6):1975–85. doi: 10.2353/ajpath.2006.051316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Lee YS, Lehar A, Sebald S, Liu M, Swaggart KA, Talbot CC, Jr, et al. Muscle hypertrophy induced by myostatin inhibition accelerates degeneration in dysferlinopathy. Hum Mol Genet. 2015 Oct 15;24(20):5711–9. doi: 10.1093/hmg/ddv288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Cohn RD, Liang HY, Shetty R, Abraham T, Wagner KR. Myostatin does not regulate cardiac hypertrophy or fibrosis. Neuromuscul Disord. 2007 Apr;17(4):290–6. doi: 10.1016/j.nmd.2007.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Bo Li Z, Zhang J, Wagner KR. Inhibition of myostatin reverses muscle fibrosis through apoptosis. J Cell Sci. 2012 Sep 1;125(Pt 17):3957–65. doi: 10.1242/jcs.090365. [DOI] [PubMed] [Google Scholar]
  • 176.Relizani K, Mouisel E, Giannesini B, Hourde C, Patel K, Morales Gonzalez S, et al. Blockade of ActRIIB signaling triggers muscle fatigability and metabolic myopathy. Mol Ther. 2014 Aug;22(8):1423–33. doi: 10.1038/mt.2014.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Quiat D, Olson EN. MicroRNAs in cardiovascular disease: from pathogenesis to prevention and treatment. J Clin Invest. 2013 Jan;123(1):11–8. doi: 10.1172/JCI62876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Crippa S, Cassano M, Messina G, Galli D, Galvez BG, Curk T, et al. miR669a and miR669q prevent skeletal muscle differentiation in postnatal cardiac progenitors. J Cell Biol. 2011 Jun 27;193(7):1197–212. doi: 10.1083/jcb.201011099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Quattrocelli M, Crippa S, Montecchiani C, Camps J, Cornaglia AI, Boldrin L, et al. Long-term miR-669a therapy alleviates chronic dilated cardiomyopathy in dystrophic mice. Journal of the American Heart Association. 2013 Aug;2(4):e000284. doi: 10.1161/JAHA.113.000284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Alloush J, Weisleder N. TRIM proteins in therapeutic membrane repair of muscular dystrophy. JAMA neurology. 2013 Jul;70(7):928–31. doi: 10.1001/jamaneurol.2013.469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Cai C, Masumiya H, Weisleder N, Matsuda N, Nishi M, Hwang M, et al. MG53 nucleates assembly of cell membrane repair machinery. Nat Cell Biol. 2009 Jan;11(1):56–64. doi: 10.1038/ncb1812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.He B, Tang RH, Weisleder N, Xiao B, Yuan Z, Cai C, et al. Enhancing Muscle Membrane Repair by Gene Delivery of MG53 Ameliorates Muscular Dystrophy and Heart Failure in delta-Sarcoglycan-deficient Hamsters. Mol Ther. 2012 Feb 7; doi: 10.1038/mt.2012.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Song R, Peng W, Zhang Y, Lv F, Wu HK, Guo J, et al. Central role of E3 ubiquitin ligase MG53 in insulin resistance and metabolic disorders. Nature. 2013 Feb 21;494(7437):375–9. doi: 10.1038/nature11834. [DOI] [PubMed] [Google Scholar]
  • 184.Yi JS, Park JS, Ham YM, Nguyen N, Lee NR, Hong J, et al. MG53-induced IRS-1 ubiquitination negatively regulates skeletal myogenesis and insulin signalling. Nat Commun. 2013;4:2354. doi: 10.1038/ncomms3354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Liu F, Song R, Feng Y, Guo J, Chen Y, Zhang Y, et al. Upregulation of MG53 induces diabetic cardiomyopathy through transcriptional activation of peroxisome proliferation-activated receptor alpha. Circulation. 2015 Mar 3;131(9):795–804. doi: 10.1161/CIRCULATIONAHA.114.012285. [DOI] [PubMed] [Google Scholar]
  • 186.Hoshijima M, Knoll R, Pashmforoush M, Chien KR. Reversal of calcium cycling defects in advanced heart failure toward molecular therapy. J Am Coll Cardiol. 2006 Nov 7;48(9 Suppl 1):A15–23. doi: 10.1016/j.jacc.2006.06.070. [DOI] [PubMed] [Google Scholar]
  • 187.Hikoso S, Ikeda Y, Yamaguchi O, Takeda T, Higuchi Y, Hirotani S, et al. Progression of heart failure was suppressed by inhibition of apoptosis signal-regulating kinase 1 via transcoronary gene transfer. J Am Coll Cardiol. 2007 Jul 31;50(5):453–62. doi: 10.1016/j.jacc.2007.03.053. [DOI] [PubMed] [Google Scholar]
  • 188.Carboni N, Marrosu G, Porcu M, Mateddu A, Solla E, Cocco E, et al. Dilated cardiomyopathy with conduction defects in a patient with partial merosin deficiency due to mutations in the laminin-alpha2-chain gene: a chance association or a novel phenotype? Muscle Nerve. 2011 Nov;44(5):826–8. doi: 10.1002/mus.22228. [DOI] [PubMed] [Google Scholar]
  • 189.Spyrou N, Philpot J, Foale R, Camici PG, Muntoni F. Evidence of left ventricular dysfunction in children with merosin-deficient congenital muscular dystrophy. Am Heart J. 1998 Sep;136(3):474–6. doi: 10.1016/s0002-8703(98)70222-4. [DOI] [PubMed] [Google Scholar]
  • 190.Marques J, Duarte ST, Costa S, Jacinto S, Oliveira J, Oliveira ME, et al. Atypical phenotype in two patients with LAMA2 mutations. Neuromuscul Disord. 2014 May;24(5):419–24. doi: 10.1016/j.nmd.2014.01.004. [DOI] [PubMed] [Google Scholar]
  • 191.Rash SM, Wanitkin S, Shiota T, Sahn DJ, Pillers DM. Congenital muscular dystrophy mouse model dy/dy has hypertrophic cardiomyopathy by echocardiography. Pediatr Res. 1998;43:26. [Google Scholar]
  • 192.Qiao C, Li J, Zhu T, Draviam R, Watkins S, Ye X, et al. Amelioration of laminin-alpha2-deficient congenital muscular dystrophy by somatic gene transfer of miniagrin. Proc Natl Acad Sci U S A. 2005 Aug 23;102(34):11999–2004. doi: 10.1073/pnas.0502137102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Vannoy CH, Xu L, Keramaris E, Lu P, Xiao X, Lu QL. Adeno-associated virus-mediated overexpression of LARGE rescues alpha-dystroglycan function in dystrophic mice with mutations in the fukutin-related protein. Human gene therapy methods. 2014 Jun;25(3):187–96. doi: 10.1089/hgtb.2013.151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194**.Duan D. Challenges and opportunities in dystrophin-deficient cardiomyopathy gene therapy. Hum Mol Genet. 2006 Oct 15;15(2):R253–61. doi: 10.1093/hmg/ddl180. This is the first comprehensive review on dystrophic cardiomyopathy gene therapy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Lai Y, Duan D. Progress in gene therapy of dystrophic heart disease. Gene Ther. 2012 Feb 9;19(6):678–85. doi: 10.1038/gt.2012.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Shin J-H, Bostick B, Yue Y, Duan D. Duchenne cardiomyopathy gene therapy. In: Duan D, editor. Muscle gene therapy. New York: Springer Science + Business Media, LLC; 2010. pp. 141–62. [Google Scholar]
  • 197.Yue Y, Skimming JW, Liu M, Strawn T, Duan D. Full-length dystrophin expression in half of the heart cells ameliorates beta-isoproterenol-induced cardiomyopathy in mdx mice. Hum Mol Genet. 2004 Aug 1;13(15):1669–75. doi: 10.1093/hmg/ddh174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Townsend D, Yasuda S, Li S, Chamberlain JS, Metzger JM. Emergent dilated cardiomyopathy caused by targeted repair of dystrophic skeletal muscle. Mol Ther. 2008 May;16(5):832–5. doi: 10.1038/mt.2008.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Wasala NB, Bostick B, Yue Y, Duan D. Exclusive skeletal muscle correction does not modulate dystrophic heart disease in the aged mdx model of Duchenne cardiomyopathy. Hum Mol Genet. 2013 Jul 1;22(13):2634–41. doi: 10.1093/hmg/ddt112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Mourkioti F, Kustan J, Kraft P, Day JW, Zhao MM, Kost-Alimova M, et al. Role of telomere dysfunction in cardiac failure in Duchenne muscular dystrophy. Nat Cell Biol. 2013 Aug;15(8):895–904. doi: 10.1038/ncb2790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Deconinck AE, Rafael JA, Skinner JA, Brown SC, Potter AC, Metzinger L, et al. Utrophin-dystrophin-deficient mice as a model for Duchenne muscular dystrophy. Cell. 1997;90(4):717–27. doi: 10.1016/s0092-8674(00)80532-2. [DOI] [PubMed] [Google Scholar]
  • 202.Grady RM, Teng H, Nichol MC, Cunningham JC, Wilkinson RS, Sanes JR. Skeletal and cardiac myopathies in mice lacking utrophin and dystrophin: a model for Duchenne muscular dystrophy. Cell. 1997;90(4):729–38. doi: 10.1016/s0092-8674(00)80533-4. [DOI] [PubMed] [Google Scholar]
  • 203.Megeney LA, Kablar B, Perry RL, Ying C, May L, Rudnicki MA. Severe cardiomyopathy in mice lacking dystrophin and MyoD. Proc Natl Acad Sci U S A. 1999;96(1):220–5. doi: 10.1073/pnas.96.1.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Chandrasekharan K, Yoon JH, Xu Y, deVries S, Camboni M, Janssen PM, et al. A human-specific deletion in mouse Cmah increases disease severity in the mdx model of Duchenne muscular dystrophy. Sci Transl Med. 2010 Jul 28;2(42):42ra54. doi: 10.1126/scitranslmed.3000692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205*.McGreevy JW, Hakim CH, McIntosh MA, Duan D. Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis Model Mech. 2015 Mar;8(3):195–213. doi: 10.1242/dmm.018424. This is a comprehensive review on animal models for Duchenne muscular dystrophy. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Smith BF, Kornegay JN, Duan D. Independent canine models of Duchenne muscular dystrophy due to intronic insertions of repetitive DNA. Mol Ther. 2007;15(Supplement 1):S51. [Google Scholar]
  • 207.Smith BF, Wrighten R. Animal models for inherited muscle diseases. In: Duan D, editor. Muscle gene therapy. New York: Springer Science + Business Media, LLC; 2010. pp. 1–21. [Google Scholar]
  • 208.Smith BF, Yue Y, Woods PR, Kornegay JN, Shin JH, Williams RR, et al. An intronic LINE-1 element insertion in the dystrophin gene aborts dystrophin expression and results in Duchenne-like muscular dystrophy in the corgi breed. Lab Invest. 2011 Feb;91(2):216–31. doi: 10.1038/labinvest.2010.146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Walmsley GL, Arechavala-Gomeza V, Fernandez-Fuente M, Burke MM, Nagel N, Holder A, et al. A Duchenne muscular dystrophy gene hot spot mutation in dystrophin-deficient cavalier king charles spaniels is amenable to exon 51 skipping. PLoS One. 2010;5(1):e8647. doi: 10.1371/journal.pone.0008647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Pan X, Yue Y, Zhang K, Lostal W, Shin JH, Duan D. Long-term robust myocardial transduction of the dog heart from a peripheral vein by adeno-associated virus serotype-8. Hum Gene Ther. 2013 Jun;24(6):584–94. doi: 10.1089/hum.2013.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Pan X, Yue Y, Zhang K, Hakim CH, Kodippili K, McDonald T, et al. AAV-8 is more efficient than AAV-9 in transducing neonatal dog heart. Human gene therapy methods. 2015;26(4):54–61. doi: 10.1089/hgtb.2014.128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zhang Y, King OD, Rahimov F, Jones TI, Ward CW, Kerr JP, et al. Human skeletal muscle xenograft as a new preclinical model for muscle disorders. Hum Mol Genet. 2014 Jun 15;23(12):3180–8. doi: 10.1093/hmg/ddu028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Lisowski L, Dane AP, Chu K, Zhang Y, Cunningham SC, Wilson EM, et al. Selection and evaluation of clinically relevant AAV variants in a xenograft liver model. Nature. 2014 Feb 20;506(7488):382–6. doi: 10.1038/nature12875. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Chen Y, Zheng Y, Kang Y, Yang W, Niu Y, Guo X, et al. Functional disruption of the dystrophin gene in rhesus monkey using CRISPR/Cas9. Hum Mol Genet. 2015 Jul 1;24(13):3764–74. doi: 10.1093/hmg/ddv120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Duan D. Duchenne muscular dystrophy gene therapy: lost in translation? Res Rep Biol. 2011;2:31–42. doi: 10.2147/RRB.S13463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Duan D. Duchenne muscular dystrophy gene therapy in the canine model. Hum Gene Ther Clin Dev. 2015 Mar;26(1):57–69. doi: 10.1089/humc.2015.006. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES