Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Hum Gene Ther ; 34(9-10): 459-470, 2023 05.
Article in English | MEDLINE | ID: mdl-36310439

ABSTRACT

Duchenne muscular dystrophy (DMD) is a fatal muscle disease caused by dystrophin deficiency. Dystrophin consists of the amino terminus, central rod domain with 24 spectrin-like repeats and four hinges (H), cysteine-rich domain, and carboxyl terminus. Several highly abbreviated micro-dystrophins (µDys) are currently in clinical trials. They all carry H1 and H4. In this study, we investigated whether these two hinges are essential for µDy function in murine DMD models. Three otherwise identical µDys were engineered to contain H1 and/or H4 and were named H1/H4 (with both H1 and H4), ΔH1 (without H1), and ΔH4 (without H4). These constructs were packaged in adeno-associated virus serotype-9 and delivered to the tibialis anterior muscle of 3-month-old male mdx4cv mice (1E12 vector genome particles/muscle). Three months later, we detected equivalent µDys expression in total muscle lysate. However, only H1/H4 and ΔH1 showed correct sarcolemmal localization. ΔH4 mainly existed as sarcoplasmic aggregates. H1/H4 and ΔH1, but not ΔH4, fully restored the dystrophin-associated protein complex and significantly improved the specific muscle force. Eccentric contraction-induced force decline was best protected by H1/H4, followed by ΔH1, but not by ΔH4. Next, we compared H1/H4 and ΔH1 in 6-week-old male mdx mice by intravenous injection (1E13 vector genome particles/mouse). Four months postinjection, H1/H4 significantly outperformed ΔH1 in extensor digitorum longus muscle force measurements but two constructs yielded comparable electrocardiography improvements. We conclude that H4 is essential for µDys function and H1 facilitates force production. Our findings will help develop next-generation µDys gene therapy.


Subject(s)
Muscular Dystrophy, Duchenne , Male , Mice , Animals , Muscular Dystrophy, Duchenne/genetics , Dystrophin/genetics , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Genetic Therapy
2.
Hum Gene Ther ; 34(9-10): 449-458, 2023 05.
Article in English | MEDLINE | ID: mdl-36515166

ABSTRACT

Adeno-associated virus (AAV)-mediated systemic micro-dystrophin (µDys) therapy is currently in clinical trials. The hope is to permanently improve the life quality of Duchenne muscular dystrophy (DMD) patients. Numerous preclinical studies have been conducted to support these trials. However, none examined whether a single therapy at a young age can lead to lifelong disease amelioration. To address this critical question, we injected 1 × 1013 vg particles/mouse of an AAV serotype-9 µDys vector to 3-month-old mdx mice through the tail vein. Therapeutic outcomes were evaluated at the age of 11 months (adulthood, 8 months postinjection) and 21 months (terminal age, 18 months postinjection). Immunostaining and Western blot showed saturated supraphysiological levels of µDys expression in skeletal muscle and heart till the end of the study. Treatment significantly improved grip force and treadmill running, and significantly reduced the serum creatine kinase level at both time points. Since cardiac death is a major threat in late-stage patients, we evaluated cardiac electrophysiology and hemodynamics by ECG and the closed-chest cardiac catheter assay, respectively. Significant improvements were observed in these assays. Importantly, many ECG and hemodynamic parameters (heart rate, PR interval, QRS duration, QTc interval, end-diastolic/systolic volume, dP/dt max and min, max pressure, and ejection fraction) were completely normalized at 21 months of age. Our results have provided direct evidence that a single systemic AAV µDys therapy has the potential to provide lifelong benefits in the murine DMD model.


Subject(s)
Muscular Dystrophy, Duchenne , Mice , Animals , Muscular Dystrophy, Duchenne/genetics , Dystrophin/genetics , Dependovirus/genetics , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Genetic Therapy/methods
3.
Hum Gene Ther ; 33(9-10): 518-528, 2022 05.
Article in English | MEDLINE | ID: mdl-35350865

ABSTRACT

Adeno-associated virus (AAV)-mediated clustered regularly interspaced short palindromic repeats (CRISPR) editing holds promise to restore missing dystrophin in Duchenne muscular dystrophy (DMD). Intramuscular coinjection of CRISPR-associated protein 9 (Cas9) and guide RNA (gRNA) vectors resulted in robust dystrophin restoration in short-term studies in the mdx mouse model of DMD. Intriguingly, this strategy failed to yield efficient dystrophin rescue in muscle in a long-term (18-month) systemic injection study. In-depth analyses revealed a selective loss of the gRNA vector after long-term systemic, but not short-term local injection. To determine whether preferential gRNA vector depletion is due to the mode of delivery (local vs. systemic) or the duration of the study (short term vs. long term), we conducted a short-term systemic injection study. The gRNA (4e12 vg/mouse in the 1:1 group or 1.2e13 vg/mouse in the 3:1 group) and Cas9 (4e12 vg/mouse) vectors were coinjected intravenously into 4-week-old mdx mice. The ratio of the gRNA to Cas9 vector genome copy dropped from 1:1 and 3:1 at injection to 0.4:1 and 1:1 at harvest 3 months later, suggesting that the route of administration, rather than the experimental duration, determines preferential gRNA vector loss. Consistent with our long-term systemic injection study, the vector ratio did not influence Cas9 expression. However, the 3:1 group showed significantly higher dystrophin expression and genome editing, better myofiber size distribution, and a more pronounced improvement in muscle function and electrocardiography. Our data suggest that the gRNA vector dose determines the outcome of systemic AAV CRISPR therapy for DMD.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Dependovirus/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Gene Editing/methods , Genetic Therapy/methods , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
4.
Nat Commun ; 12(1): 6769, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819506

ABSTRACT

Adeno-associated virus (AAV)-mediated CRISPR-Cas9 editing holds promise to treat many diseases. The immune response to bacterial-derived Cas9 has been speculated as a hurdle for AAV-CRISPR therapy. However, immunological consequences of AAV-mediated Cas9 expression have thus far not been thoroughly investigated in large mammals. We evaluate Cas9-specific immune responses in canine models of Duchenne muscular dystrophy (DMD) following intramuscular and intravenous AAV-CRISPR therapy. Treatment results initially in robust dystrophin restoration in affected dogs but also induces muscle inflammation, and Cas9-specific humoral and cytotoxic T-lymphocyte (CTL) responses that are not prevented by the muscle-specific promoter and transient prednisolone immune suppression. In normal dogs, AAV-mediated Cas9 expression induces similar, though milder, immune responses. In contrast, other therapeutic (micro-dystrophin and SERCA2a) and reporter (alkaline phosphatase, AP) vectors result in persistent expression without inducing muscle inflammation. Our results suggest Cas9 immunity may represent a critical barrier for AAV-CRISPR therapy in large mammals.


Subject(s)
CRISPR-Cas Systems/immunology , Genetic Therapy/adverse effects , Genetic Vectors/immunology , Muscle, Skeletal/immunology , Muscular Dystrophy, Duchenne/therapy , Animals , CRISPR-Cas Systems/genetics , Dependovirus/genetics , Disease Models, Animal , Dogs , Dystrophin/genetics , Dystrophin/immunology , Gene Editing/methods , Genes, Reporter/genetics , Genes, Reporter/immunology , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Humans , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/immunology
5.
Hum Mol Genet ; 30(14): 1321-1336, 2021 06 26.
Article in English | MEDLINE | ID: mdl-33949649

ABSTRACT

ΔR4-R23/ΔCT micro-dystrophin (µDys) is a miniaturized version of dystrophin currently evaluated in a Duchenne muscular dystrophy (DMD) gene therapy trial to treat skeletal and cardiac muscle disease. In pre-clinical studies, µDys efficiently rescues cardiac histopathology, but only partially normalizes cardiac function. To gain insights into factors that may impact the cardiac therapeutic efficacy of µDys, we compared by mass spectrometry the composition of purified dystrophin and µDys protein complexes in the mouse heart. We report that compared to dystrophin, µDys has altered associations with α1- and ß2-syntrophins, as well as cavins, a group of caveolae-associated signaling proteins. In particular, we found that membrane localization of cavin-1 and cavin-4 in cardiomyocytes requires dystrophin and is profoundly disrupted in the heart of mdx5cv mice, a model of DMD. Following cardiac stress/damage, membrane-associated cavin-4 recruits the signaling molecule ERK to caveolae, which activates key cardio-protective responses. Evaluation of ERK signaling revealed a profound inhibition, below physiological baseline, in the mdx5cv mouse heart. Expression of µDys in mdx5cv mice prevented the development of cardiac histopathology but did not rescue membrane localization of cavins nor did it normalize ERK signaling. Our study provides the first comparative analysis of purified protein complexes assembled in vivo by full-length dystrophin and a therapeutic micro-dystrophin construct. This has revealed disruptions in cavins and ERK signaling that may contribute to DMD cardiomyopathy. This new knowledge is important for ongoing efforts to prevent and treat heart disease in DMD patients.


Subject(s)
Cardiomyopathies , Muscular Dystrophy, Duchenne , Animals , Cardiomyopathies/genetics , Dystrophin/metabolism , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/metabolism , Myocytes, Cardiac/metabolism , Proteomics
6.
Expert Opin Drug Discov ; 15(4): 443-456, 2020 04.
Article in English | MEDLINE | ID: mdl-32000537

ABSTRACT

Introduction: Duchenne muscular dystrophy (DMD) is an X-linked handicapping disease due to the loss of an essential muscle protein dystrophin. Dystrophin-null animals have been extensively used to study disease mechanisms and to develop experimental therapeutics. Despite decades of research, however, treatment options for DMD remain very limited.Areas covered: High-throughput high-content screening and precision medicine offer exciting new opportunities. Here, the authors review animal models that are suitable for these studies.Expert opinion: Nonmammalian models (worm, fruit fly, and zebrafish) are particularly attractive for cost-effective large-scale drug screening. Several promising lead compounds have been discovered using these models. Precision medicine for DMD aims at developing mutation-specific therapies such as exon-skipping and genome editing. To meet these needs, models with patient-like mutations have been established in different species. Models that harbor hotspot mutations are very attractive because the drugs developed in these models can bring mutation-specific therapies to a large population of patients. Humanized hDMD mice carry the entire human dystrophin gene in the mouse genome. Reagents developed in the hDMD mouse-based models are directly translatable to human patients.


Subject(s)
Disease Models, Animal , Drug Discovery/methods , Muscular Dystrophy, Duchenne/drug therapy , Animals , Drug Development , Dystrophin/genetics , High-Throughput Screening Assays/methods , Humans , Mice , Muscular Dystrophy, Duchenne/genetics , Mutation , Precision Medicine
7.
Mol Ther ; 28(3): 845-854, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31981493

ABSTRACT

Loss of dystrophin leads to Duchenne muscular dystrophy (DMD). A pathogenic feature of DMD is the significant elevation of cytosolic calcium. Supraphysiological calcium triggers protein degradation, membrane damage, and eventually muscle death and dysfunction. Sarcoplasmic/endoplasmic reticulum (SR) calcium ATPase (SERCA) is a calcium pump that transports cytosolic calcium to the SR during excitation-contraction coupling. We hypothesize that a single systemic delivery of SERCA2a with adeno-associated virus (AAV) may improve calcium recycling and provide long-lasting benefits in DMD. To test this, we injected an AAV9 human SERCA2a vector (6 × 1012 viral genome particles/mouse) intravenously to 3-month-old mdx mice, the most commonly used DMD model. Immunostaining and western blot showed robust human SERCA2a expression in the heart and skeletal muscle for 18 months. Concomitantly, SR calcium uptake was significantly improved in these tissues. SERCA2a therapy significantly enhanced grip force and treadmill performance, completely prevented myocardial fibrosis, and normalized electrocardiograms (ECGs). Cardiac catheterization showed normalization of multiple systolic and diastolic hemodynamic parameters in treated mice. Importantly, chamber dilation was completely prevented, and ejection fraction was restored to the wild-type level. Our results suggest that a single systemic AAV9 SERCA2a therapy has the potential to provide long-lasting benefits for DMD.


Subject(s)
Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/therapy , Gene Expression , Genetic Therapy , Muscular Dystrophy, Duchenne/complications , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Administration, Intravenous , Animals , Dependovirus/genetics , Disease Models, Animal , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Sarcoplasmic Reticulum/metabolism , Time Factors , Transduction, Genetic
8.
Mol Ther ; 27(9): 1568-1585, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31327755

ABSTRACT

CRISPR editing of muscle stem cells (MuSCs) with adeno-associated virus serotype-9 (AAV9) holds promise for sustained gene repair therapy for muscular dystrophies. However, conflicting evidence exists on whether AAV9 transduces MuSCs. To rigorously address this question, we used a muscle graft model. The grafted muscle underwent complete necrosis before regenerating from its MuSCs. We injected AAV9.Cre into Ai14 mice. These mice express tdTomato upon Cre-mediated removal of a floxed stop codon. About 28%-47% and 24%-89% of Pax7+ MuSCs expressed tdTomato in pre-grafts and regenerated grafts (p > 0.05), respectively, suggesting AAV9 efficiently transduced MuSCs, and AAV9-edited MuSCs renewed successfully. Robust MuSC transduction was further confirmed by delivering AAV9.Cre to Pax7-ZsGreen-Ai14 mice in which Pax7+ MuSCs are genetically labeled by ZsGreen. Next, we co-injected AAV9.Cas9 and AAV9.gRNA to dystrophic mdx mice to repair the mutated dystrophin gene. CRISPR-treated and untreated muscles were grafted to immune-deficient, dystrophin-null NSG.mdx4cv mice. Grafts regenerated from CRISPR-treated muscle contained the edited genome and yielded 2.7-fold more dystrophin+ cells (p = 0.015). Importantly, increased dystrophin expression was not due to enhanced formation of revertant fibers or de novo transduction by residual CRISPR vectors in the graft. We conclude that AAV9 effectively transduces MuSCs. AAV9 CRISPR editing of MuSCs may provide enduring therapy.


Subject(s)
Dependovirus/genetics , Dystrophin/genetics , Gene Editing , Genetic Vectors/genetics , Myoblasts/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Disease Models, Animal , Dystrophin/chemistry , Gene Expression , Gene Transfer Techniques , Genes, Reporter , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , RNA, Guide, Kinetoplastida/genetics , Regeneration , Satellite Cells, Skeletal Muscle/metabolism , Transduction, Genetic
9.
Hum Gene Ther ; 30(5): 535-543, 2019 05.
Article in English | MEDLINE | ID: mdl-30648435

ABSTRACT

Clustered regularly interspaced short palindromic repeats (CRISPR) editing is being considered as a potential gene repair therapy to treat Duchenne muscular dystrophy, a dystrophin-deficient lethal muscle disease affecting all muscles in the body. A recent preliminary study from the Olson laboratory (Amoasii et al. Science 2018;362:89-91) showed robust dystrophin restoration in a canine Duchenne muscular dystrophy model following intramuscular or intravenous delivery of the CRISPR editing machinery by adeno-associated virus serotype 9. Despite the limitation of the small sample size, short study duration, and the lack of muscle function data, the Olson lab findings have provided important proof of principle for scaling up CRISPR therapy from rodents to large mammals. Future large-scale, long-term, and comprehensive studies are warranted to establish the safety and efficacy of CRISPR editing therapy in large mammals.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Muscular Dystrophy, Duchenne/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dogs , Genetic Vectors/genetics , Humans , Muscular Dystrophy, Duchenne/therapy , RNA, Guide, Kinetoplastida
10.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518686

ABSTRACT

Adeno-associated virus-mediated (AAV-mediated) CRISPR editing is a revolutionary approach for treating inherited diseases. Sustained, often life-long mutation correction is required for treating these diseases. Unfortunately, this has never been demonstrated with AAV CRISPR therapy. We addressed this question in the mdx model of Duchenne muscular dystrophy (DMD). DMD is caused by dystrophin gene mutation. Dystrophin deficiency leads to ambulation loss and cardiomyopathy. We treated 6-week-old mice intravenously and evaluated disease rescue at 18 months. Surprisingly, nominal dystrophin was restored in skeletal muscle. Cardiac dystrophin was restored, but histology and hemodynamics were not improved. To determine the underlying mechanism, we evaluated components of the CRISPR-editing machinery. Intriguingly, we found disproportional guide RNA (gRNA) vector depletion. To test whether this is responsible for the poor outcome, we increased the gRNA vector dose and repeated the study. This strategy significantly increased dystrophin restoration and reduced fibrosis in all striated muscles at 18 months. Importantly, skeletal muscle function and cardiac hemodynamics were significantly enhanced. Interestingly, we did not see selective depletion of the gRNA vector after intramuscular injection. Our results suggest that gRNA vector loss is a unique barrier for systemic AAV CRISPR therapy. This can be circumvented by vector dose optimization.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Dystrophin/genetics , Gene Editing , Muscular Dystrophy, Duchenne/genetics , Animals , Dependovirus , Disease Models, Animal , Female , Fibrosis , Genetic Therapy , Genetic Vectors , Male , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Mutation , Myocardium/metabolism , Myocardium/pathology , Neuromuscular Diseases , RNA, Guide, Kinetoplastida
11.
Hum Gene Ther ; 29(7): 737-748, 2018 07.
Article in English | MEDLINE | ID: mdl-29433343

ABSTRACT

Heart disease is a major health threat for Duchenne/Becker muscular dystrophy patients and carriers. Expression of a 6-8 kb mini-dystrophin gene in the heart holds promise to change the disease course dramatically. However, the mini-dystrophin gene cannot be easily studied with adeno-associated virus (AAV) gene delivery because the size of the minigene exceeds AAV packaging capacity. Cardiac protection of the ΔH2-R19 minigene was previously studied using the cardiac-specific transgenic approach. Although this minigene fully normalized skeletal muscle force, it only partially corrected electrocardiogram and heart hemodynamics in dystrophin-null mdx mice that had moderate cardiomyopathy. This study evaluated the ΔH2-R15 minigene using the same transgenic approach in mdx mice that had more severe cardiomyopathy. In contrast to the ΔH2-R19 minigene, the ΔH2-R15 minigene carries dystrophin spectrin-like repeats 16 to 19 (R16-19), a region that has been suggested to protect the heart in clinical studies. Cardiac expression of the ΔH2-R15 minigene normalized all aberrant electrocardiogram changes and improved hemodynamics. Importantly, it corrected the end-diastolic volume, an important diastolic parameter not rescued by ΔH2-R19 mini-dystrophin. It is concluded that that ΔH2-R15 mini-dystrophin is a superior candidate gene for heart protection. This finding has important implications in the design of the mini/micro-dystrophin gene for Duchenne cardiomyopathy therapy.


Subject(s)
Cardiomyopathy, Dilated/therapy , Dystrophin/genetics , Genetic Therapy , Myocardium/metabolism , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Dystrophin/administration & dosage , Dystrophin/biosynthesis , Electrocardiography , Gene Expression Regulation/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Humans , Mice , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Myocardium/pathology , Stroke Volume/genetics
12.
Mol Ther Methods Clin Dev ; 6: 216-230, 2017 Sep 15.
Article in English | MEDLINE | ID: mdl-28932757

ABSTRACT

Micro-dystrophins are highly promising candidates for treating Duchenne muscular dystrophy, a lethal muscle disease caused by dystrophin deficiency. Here, we report robust disease rescue in the severe DBA/2J-mdx model with a neuronal nitric oxide synthase (nNOS)-binding micro-dystrophin vector. 2 × 1013 vector genome particles/mouse of the vector were delivered intravenously to 10-week-old mice and were evaluated at 6 months of age. Saturated micro-dystrophin expression was detected in all skeletal muscles and the heart and restored the dystrophin-associated glycoprotein complex and nNOS. In skeletal muscle, therapy substantially reduced fibrosis and calcification and significantly attenuated inflammation. Centronucleation was significantly decreased in the tibialis anterior (TA) and extensor digitorum longus (EDL) muscles but not in the quadriceps. Muscle function was normalized in the TA and significantly improved in the EDL muscle. Heart histology and function were also evaluated. Consistent with the literature, DBA/2J-mdx mice showed myocardial calcification and fibrosis and cardiac hemodynamics was compromised. Surprisingly, similar myocardial pathology and hemodynamic defects were detected in control DBA/2J mice. As a result, interpretation of the cardiac data proved difficult due to the confounding phenotype in control DBA/2J mice. Our results support further development of this microgene vector for clinical translation. Further, DBA/2J-mdx mice are not good models for Duchenne cardiomyopathy.

13.
J Mol Cell Cardiol ; 102: 45-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27908661

ABSTRACT

Dystrophin deficiency results in Duchenne cardiomyopathy, a primary cause of death in Duchenne muscular dystrophy (DMD). Gene therapy has shown great promise in ameliorating the cardiac phenotype in mouse models of DMD. However, it is not completely clear how much dystrophin is required to treat dystrophic heart disease. We and others have shown that mosaic dystrophin expression at the wild-type level, depending on the percentage of dystrophin positive cardiomyocytes, can either delay the onset of or fully prevent cardiomyopathy in dystrophin-null mdx mice. Many gene therapy strategies will unlikely restore dystrophin to the wild-type level in a cardiomyocyte. To determine whether low-level dystrophin expression can reduce the cardiac manifestations in DMD, we examined heart histology, ECG and hemodynamics in 21-m-old normal BL6 and two strains of BL6-background dystrophin-deficient mice. Mdx3cv mice show uniform low-level expression of a near full-length dystrophin protein in every myofiber while mdx4cv mice have no dystrophin expression. Immunostaining and western blot confirmed marginal level dystrophin expression in the heart of mdx3cv mice. Although low-level expression did not reduce myocardial histopathology, it significantly ameliorated QRS prolongation and normalized diastolic hemodynamic deficiencies. Our study demonstrates for the first time that low-level dystrophin can partially preserve heart function.


Subject(s)
Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Dystrophin/genetics , Gene Expression , Muscular Dystrophy, Duchenne/complications , Myocardium/metabolism , Age Factors , Animals , Biomarkers , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calsequestrin/genetics , Calsequestrin/metabolism , Cardiomyopathies/diagnosis , Diastole , Disease Models, Animal , Dystrophin/metabolism , Electrocardiography , Mice , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Ventricular Function
14.
Mol Ther Methods Clin Dev ; 3: 16045, 2016.
Article in English | MEDLINE | ID: mdl-27419194

ABSTRACT

Dystrophin gene replacement holds the promise of treating Duchenne muscular dystrophy. Supraphysiological expression is a concern for all gene therapy studies. In the case of Duchenne muscular dystrophy, Chamberlain and colleagues found that 50-fold overexpression did not cause deleterious side effect in skeletal muscle. To determine whether excessive dystrophin expression in the heart is safe, we studied two lines of transgenic mdx mice that selectively expressed a therapeutic minidystrophin gene in the heart at 50-fold and 100-fold of the normal levels. In the line with 50-fold overexpression, minidystrophin showed sarcolemmal localization and electrocardiogram abnormalities were corrected. However, in the line with 100-fold overexpression, we not only detected sarcolemmal minidystrophin expression but also observed accumulation of minidystrophin vesicles in the sarcoplasm. Excessive minidystrophin expression did not correct tachycardia, a characteristic feature of Duchenne muscular dystrophy. Importantly, several electrocardiogram parameters (QT interval, QRS duration and the cardiomyopathy index) became worse than that of mdx mice. Our data suggests that the mouse heart can tolerate 50-fold minidystrophin overexpression, but 100-fold overexpression leads to cardiac toxicity.

15.
Hum Mol Genet ; 25(13): 2633-2644, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27106099

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by dystrophin deficiency. A fundamental question in DMD pathogenesis and dystrophin gene therapy is whether muscle health depends on continuous dystrophin expression throughout the life. Published data suggest that transient dystrophin expression in early life might offer permanent protection. To study the consequences of adulthood dystrophin loss, we generated two strains of floxed mini-dystrophin transgenic mice on the dystrophin-null background. Muscle diseases were prevented in skeletal muscle of the YL238 strain and the heart of the SJ13 strain by selective expression of a therapeutic mini-dystrophin gene in skeletal muscle and heart, respectively. The mini-dystrophin gene was removed from the tibialis anterior (TA) muscle of 8-month-old YL238 mice and the heart of 7-month-old SJ13 mice using an adeno-associated virus serotype-9 Cre recombinase vector (AAV.CBA.Cre). At 12 and 15 months after AAV.CBA.Cre injection, mini-dystrophin expression was reduced by ∼87% in the TA muscle of YL238 mice and ∼64% in the heart of SJ13 mice. Mini-dystrophin reduction caused muscle atrophy, degeneration and force loss in the TA muscle of YL238 mice and significantly compromised left ventricular hemodynamics in SJ13 mice. Our results suggest that persistent dystrophin expression is essential for continuous muscle and heart protection.


Subject(s)
Dystrophin/metabolism , Dystrophin/physiology , Muscular Dystrophy, Duchenne/metabolism , Animals , Dystrophin/genetics , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Genomics , Mice , Mice, Inbred CBA , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Myocardium/metabolism
16.
PLoS Curr ; 72015 Feb 10.
Article in English | MEDLINE | ID: mdl-25737807

ABSTRACT

The mdx mouse is the most frequently used animal model for Duchenne muscular dystrophy (DMD), a fatal muscle disease caused by the loss of dystrophin. Mdx mice are naturally occurring dystrophin-null mice on the C57BL/10 (BL10) background. We crossed black mdx to the white FVB background and generated mdx/FVB mice. Compared to that of age- and sex-matched FVB mice, mdx/FVB mice showed characteristic limb muscle pathology similar to that of original mdx mice. Further, the forelimb grip strength and limb muscle (tibialis anterior and extensor digitorum longus) specific force of mdx/FVB mice were significantly lower than that of wild type FVB mice. Consistent with what has been reported in original mdx mice, mdx/FVB mice also showed increased susceptibility to eccentric contraction-induced force loss and elevated serum creatine kinase. Our results suggest that the FVB background does not dramatically alter the dystrophic phenotype of mdx mice.

17.
Hum Mol Genet ; 23(12): 3189-99, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24463882

ABSTRACT

Transgenic gene deletion/over-expression studies have established the cardioprotective role of neuronal nitric oxide synthase (nNOS). However, it remains unclear whether nNOS-mediated heart protection can be translated to gene therapy. In this study, we generated an adeno-associated virus (AAV) nNOS vector and tested its therapeutic efficacy in the aged mdx model of Duchenne cardiomyopathy. A PDZ domain-deleted nNOS gene (ΔPDZ nNOS) was packaged into tyrosine mutant AAV-9 and delivered to the heart of ~14-month-old female mdx mice, a phenotypic model of Duchenne cardiomyopathy. Seven months later, we observed robust nNOS expression in the myocardium. Supra-physiological ΔPDZ nNOS expression significantly reduced myocardial fibrosis, inflammation and apoptosis. Importantly, electrocardiography and left ventricular hemodynamics were significantly improved in treated mice. Additional studies revealed increased phosphorylation of phospholamban and p70S6K. Collectively, we have demonstrated the therapeutic efficacy of the AAV ΔPDZ nNOS vector in a symptomatic Duchenne cardiomyopathy model. Our results suggest that the cardioprotective role of ΔPDZ nNOS is likely through reduced apoptosis, enhanced phospholamban phosphorylation and improved Akt/mTOR/p70S6K signaling. Our study has opened the door to treat Duchenne cardiomyopathy with ΔPDZ nNOS gene transfer.


Subject(s)
Cardiomyopathies/physiopathology , Cardiomyopathies/therapy , Dependovirus/genetics , Genetic Therapy/methods , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/therapy , Nitric Oxide Synthase Type I/genetics , Animals , Apoptosis , Calcium-Binding Proteins/metabolism , Cardiomyopathies/complications , Dependovirus/metabolism , Female , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genetic Vectors/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase Type I/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction
18.
Hum Mol Genet ; 22(13): 2634-41, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23459935

ABSTRACT

Duchenne muscular dystrophy (DMD) is characterized by severe degeneration and necrosis of both skeletal and cardiac muscle. While many experimental therapies have shown great promise in treating skeletal muscle disease, an effective therapy for Duchenne cardiomyopathy remains a challenge in large animal models and human patients. The current views on cardiac consequences of skeletal muscle-centered therapy are controversial. Studies performed in young adult mdx mice (a mild DMD mouse model) have yielded opposing results. Since mdx mice do not develop dystrophic cardiomyopathy until ≥21 months of age, we reasoned that old mdx mice may represent a better model to assess the impact of skeletal muscle rescue on dystrophic heart disease. Here, we aged skeletal muscle-specific micro-dystrophin transgenic mdx mice to 23 months and examined the cardiac phenotype. As expected, transgenic mdx mice had minimal skeletal muscle disease and they also outperformed original mdx mice on treadmill running. On cardiac examination, the dystrophin-null heart of transgenic mdx mice displayed severe cardiomyopathy matching that of non-transgenic mdx mice. Specifically, both the strains showed similar heart fibrosis and cardiac function deterioration in systole and diastole. Cardiac output and ejection fraction were also equally compromised. Our results suggest that skeletal muscle rescue neither aggravates nor alleviates cardiomyopathy in aged mdx mice. These findings underscore the importance of treating both skeletal and cardiac muscles in DMD therapy.


Subject(s)
Cardiomyopathies/etiology , Muscular Dystrophy, Duchenne/complications , Age Factors , Animals , Body Weight/genetics , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Fibrosis , Heart Ventricles/physiopathology , Male , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/metabolism , Myocardium/pathology , Organ Size/genetics
19.
J Vis Exp ; (72)2013 Feb 09.
Article in English | MEDLINE | ID: mdl-23426237

ABSTRACT

Body movements are mainly provided by mechanical function of skeletal muscle. Skeletal muscle is composed of numerous bundles of myofibers that are sheathed by intramuscular connective tissues. Each myofiber contains many myofibrils that run longitudinally along the length of the myofiber. Myofibrils are the contractile apparatus of muscle and they are composed of repeated contractile units known as sarcomeres. A sarcomere unit contains actin and myosin filaments that are spaced by the Z discs and titin protein. Mechanical function of skeletal muscle is defined by the contractile and passive properties of muscle. The contractile properties are used to characterize the amount of force generated during muscle contraction, time of force generation and time of muscle relaxation. Any factor that affects muscle contraction (such as interaction between actin and myosin filaments, homeostasis of calcium, ATP/ADP ratio, etc.) influences the contractile properties. The passive properties refer to the elastic and viscous properties (stiffness and viscosity) of the muscle in the absence of contraction. These properties are determined by the extracellular and the intracellular structural components (such as titin) and connective tissues (mainly collagen) (1-2). The contractile and passive properties are two inseparable aspects of muscle function. For example, elbow flexion is accomplished by contraction of muscles in the anterior compartment of the upper arm and passive stretch of muscles in the posterior compartment of the upper arm. To truly understand muscle function, both contractile and passive properties should be studied. The contractile and/or passive mechanical properties of muscle are often compromised in muscle diseases. A good example is Duchenne muscular dystrophy (DMD), a severe muscle wasting disease caused by dystrophin deficiency (3). Dystrophin is a cytoskeletal protein that stabilizes the muscle cell membrane (sarcolemma) during muscle contraction (4). In the absence of dystrophin, the sarcolemma is damaged by the shearing force generated during force transmission. This membrane tearing initiates a chain reaction which leads to muscle cell death and loss of contractile machinery. As a consequence, muscle force is reduced and dead myofibers are replaced by fibrotic tissues (5). This later change increases muscle stiffness (6). Accurate measurement of these changes provides important guide to evaluate disease progression and to determine therapeutic efficacy of novel gene/cell/pharmacological interventions. Here, we present two methods to evaluate both contractile and passive mechanical properties of the extensor digitorum longus (EDL) muscle and the contractile properties of the tibialis anterior (TA) muscle.


Subject(s)
Muscle, Skeletal/physiology , Animals , Mice , Muscle Contraction
20.
Vector Borne Zoonotic Dis ; 13(3): 193-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23421885

ABSTRACT

Animal models for Ehrlichia chaffeensis have been unsuccessful in recapitulating the natural disease cycle. We have developed an animal model for tick feeding and transmission using white-tailed deer (Odocoileus virgianus), the intracellular bacterium (Ehrlichia chaffeensis), and the lone star tick vector (Amblyomma americanum). Here, we report the acquisition and transmission of E. chaffeensis infections by refeeding male ticks in this experimental model. This finding is important because techniques for gene silencing are most successful for unfed adult ticks. Males are able to refeed several days after acquiring a tick-borne pathogen. Using refeeding male lone star ticks and RNA interference technology, we plan to decipher underlying molecular mechanisms involved in transmitting E. chaffeensis to a host via a lone star tick bite.


Subject(s)
Arachnid Vectors/microbiology , Deer/microbiology , Ehrlichia chaffeensis/physiology , Ehrlichiosis/transmission , Ixodidae/microbiology , Animals , Deer/parasitology , Disease Models, Animal , Ehrlichiosis/microbiology , Feeding Behavior , Humans , Male , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL