Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 27(9): 1568-1585, 2019 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-31327755

RESUMEN

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.


Asunto(s)
Dependovirus/genética , Distrofina/genética , Edición Génica , Vectores Genéticos/genética , Mioblastos/metabolismo , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Distrofina/química , Expresión Génica , Técnicas de Transferencia de Gen , Genes Reporteros , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , ARN Guía de Kinetoplastida/genética , Regeneración , Células Satélite del Músculo Esquelético/metabolismo , Transducción Genética
2.
J Physiol ; 596(21): 5199-5216, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30152022

RESUMEN

KEY POINTS: We developed a novel method to study sympatholysis in dogs. We showed abolishment of sarcolemmal nNOS, and reduction of total nNOS and total eNOS in the canine Duchenne muscular dystrophy (DMD) model. We showed sympatholysis in dogs involving both nNOS-derived NO-dependent and NO-independent mechanisms. We showed that the loss of sarcolemmal nNOS compromised sympatholysis in the canine DMD model. We showed that NO-independent sympatholysis was not affected in the canine DMD model. ABSTRACT: The absence of dystrophin in Duchenne muscular dystrophy (DMD) leads to the delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma. Sarcolemmal nNOS plays an important role in sympatholysis, a process of attenuating reflex sympathetic vasoconstriction during exercise to ensure blood perfusion in working muscle. Delocalization of nNOS compromises sympatholysis resulting in functional ischaemia and muscle damage in DMD patients and mouse models. Little is known about the contribution of membrane-associated nNOS to blood flow regulation in dystrophin-deficient DMD dogs. We tested the hypothesis that the loss of sarcolemmal nNOS abolishes protective sympatholysis in contracting muscle of affected dogs. Haemodynamic responses to noradrenaline in the brachial artery were evaluated at rest and during contraction in the absence and presence of NOS inhibitors. We found sympatholysis was significantly compromised in DMD dogs, as well as in normal dogs treated with a selective nNOS inhibitor, suggesting that the absence of sarcolemmal nNOS underlies defective sympatholysis in the canine DMD model. Surprisingly, inhibition of all NOS isoforms did not completely abolish sympatholysis in normal dogs, suggesting sympatholysis in canine muscle also involves NO-independent mechanism(s). Our study established a foundation for using the dog model to test therapies aimed at restoring nNOS homeostasis in DMD.


Asunto(s)
Distrofia Muscular de Duchenne/fisiopatología , Óxido Nítrico/metabolismo , Norepinefrina/farmacología , Vasoconstricción , Vasoconstrictores/farmacología , Animales , Arteria Braquial/efectos de los fármacos , Arteria Braquial/fisiopatología , Perros , Femenino , Masculino , Distrofia Muscular de Duchenne/metabolismo , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo I/metabolismo
3.
Hum Mol Genet ; 25(17): 3647-3653, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27378693

RESUMEN

Dystrophin is a large sub-sarcolemmal protein. Its absence leads to Duchenne muscular dystrophy (DMD). Binding to the sarcolemma is essential for dystrophin to protect muscle from contraction-induced injury. It has long been thought that membrane binding of dystrophin depends on its cysteine-rich (CR) domain. Here, we provide in vivo evidence suggesting that dystrophin contains three additional membrane-binding domains including spectrin-like repeats (R)1-3, R10-12 and C-terminus (CT). To systematically study dystrophin membrane binding, we split full-length dystrophin into ten fragments and examined subcellular localizations of each fragment by adeno-associated virus-mediated gene transfer. In skeletal muscle, R1-3, CR domain and CT were exclusively localized at the sarcolemma. R10-12 showed both cytosolic and sarcolemmal localization. Importantly, the CR-independent membrane binding was conserved in murine and canine muscles. A critical function of the CR-mediated membrane interaction is the assembly of the dystrophin-associated glycoprotein complex (DGC). While R1-3 and R10-12 did not restore the DGC, surprisingly, CT alone was sufficient to establish the DGC at the sarcolemma. Additional studies suggest that R1-3 and CT also bind to the sarcolemma in the heart, though relatively weak. Taken together, our study provides the first conclusive in vivo evidence that dystrophin contains multiple independent membrane-binding domains. These structurally and functionally distinctive membrane-binding domains provide a molecular framework for dystrophin to function as a shock absorber and signaling hub. Our results not only shed critical light on dystrophin biology and DMD pathogenesis, but also provide a foundation for rationally engineering minimized dystrophins for DMD gene therapy.


Asunto(s)
Distrofina/química , Distrofina/metabolismo , Glicoproteínas/metabolismo , Distrofia Muscular Animal/metabolismo , Miocardio/metabolismo , Animales , Sitios de Unión , Secuencia Conservada , Citosol/metabolismo , Perros , Distrofina/genética , Humanos , Ratones , Ratones Endogámicos mdx , Dominios Proteicos , Sarcolema/metabolismo
4.
Hum Mol Genet ; 24(20): 5880-90, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26264580

RESUMEN

The ultimate goal of muscular dystrophy gene therapy is to treat all muscles in the body. Global gene delivery was demonstrated in dystrophic mice more than a decade ago using adeno-associated virus (AAV). However, translation to affected large mammals has been challenging. The only reported attempt was performed in newborn Duchenne muscular dystrophy (DMD) dogs. Unfortunately, AAV injection resulted in growth delay, muscle atrophy and contracture. Here we report safe and bodywide AAV delivery in juvenile DMD dogs. Three ∼2-m-old affected dogs received intravenous injection of a tyrosine-engineered AAV-9 reporter or micro-dystrophin (µDys) vector at the doses of 1.92-6.24 × 10(14) viral genome particles/kg under transient or sustained immune suppression. DMD dogs tolerated injection well and their growth was not altered. Hematology and blood biochemistry were unremarkable. No adverse reactions were observed. Widespread muscle transduction was seen in skeletal muscle, the diaphragm and heart for at least 4 months (the end of the study). Nominal expression was detected in internal organs. Improvement in muscle histology was observed in µDys-treated dogs. In summary, systemic AAV gene transfer is safe and efficient in young adult dystrophic large mammals. This may translate to bodywide gene therapy in pediatric patients in the future.


Asunto(s)
Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Transducción Genética/métodos , Administración Intravenosa , Animales , Perros , Distrofina/uso terapéutico , Femenino , Vectores Genéticos , Masculino , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/terapia
5.
Hum Mol Genet ; 22(18): 3720-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23681067

RESUMEN

Neuronal nitric oxide synthase (nNOS) membrane delocalization contributes to the pathogenesis of Duchenne muscular dystrophy (DMD) by promoting functional muscle ischemia and exacerbating muscle injury during exercise. We have previously shown that supra-physiological expression of nNOS-binding mini-dystrophin restores normal blood flow regulation and prevents functional ischemia in transgenic mdx mice, a DMD model. A critical next issue is whether systemic dual adeno-associated virus (AAV) gene therapy can restore nNOS-binding mini-dystrophin expression and mitigate muscle activity-related functional ischemia and injury. Here, we performed systemic gene transfer in mdx and mdx4cv mice using a pair of dual AAV vectors that expressed a 6 kb nNOS-binding mini-dystrophin gene. Vectors were packaged in tyrosine mutant AAV-9 and co-injected (5 × 10(12) viral genome particles/vector/mouse) via the tail vein to 1-month-old dystrophin-null mice. Four months later, we observed 30-50% mini-dystrophin positive myofibers in limb muscles. Treatment ameliorated histopathology, increased muscle force and protected against eccentric contraction-induced injury. Importantly, dual AAV therapy successfully prevented chronic exercise-induced muscle force drop. Doppler hemodynamic assay further showed that therapy attenuated adrenergic vasoconstriction in contracting muscle. Our results suggest that partial transduction can still ameliorate nNOS delocalization-associated functional deficiency. Further evaluation of nNOS binding mini-dystrophin dual AAV vectors is warranted in dystrophic dogs and eventually in human patients.


Asunto(s)
Dependovirus/genética , Distrofina/genética , Músculo Esquelético/lesiones , Distrofia Muscular de Duchenne/terapia , Óxido Nítrico Sintasa de Tipo I/metabolismo , Condicionamiento Físico Animal/efectos adversos , Animales , Perros , Distrofina/metabolismo , Terapia Genética , Vectores Genéticos , Humanos , Isquemia/terapia , Masculino , Ratones , Ratones Transgénicos , Contracción Muscular , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patología , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patología , Sarcolema/enzimología
6.
Mol Ther ; 21(4): 750-7, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23319056

RESUMEN

Dystrophin deficiency results in lethal Duchenne muscular dystrophy (DMD). Substituting missing dystrophin with abbreviated microdystrophin has dramatically alleviated disease in mouse DMD models. Unfortunately, translation of microdystrophin therapy has been unsuccessful in dystrophic dogs, the only large mammalian model. Approximately 70% of the dystrophin-coding sequence is removed in microdystrophin. Intriguingly, loss of ≥50% dystrophin frequently results in severe disease in patients. To test whether the small gene size constitutes a fundamental design error for large mammalian muscle, we performed a comprehensive study using 22 dogs (8 normal and 14 dystrophic). We delivered the ΔR2-15/ΔR18-19/ΔR20-23/ΔC microdystrophin gene to eight extensor carpi ulnaris (ECU) muscles in six dystrophic dogs using Y713F tyrosine mutant adeno-associated virus (AAV)-9 (2.6 × 10(13) viral genome (vg) particles/muscle). Robust expression was observed 2 months later despite T-cell infiltration. Major components of the dystrophin-associated glycoprotein complex (DGC) were restored by microdystrophin. Treated muscle showed less inflammation, fibrosis, and calcification. Importantly, therapy significantly preserved muscle force under the stress of repeated cycles of eccentric contraction. Our results have established the proof-of-concept for microdystrophin therapy in dystrophic muscles of large mammals and set the stage for clinical trial in human patients.


Asunto(s)
Distrofina/metabolismo , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animales , Western Blotting , Perros , Distrofina/genética , Técnicas de Transferencia de Gen , Masculino , Ratones , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/metabolismo
7.
Mol Ther Methods Clin Dev ; 32(2): 101268, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38911286

RESUMEN

Excessive cytosolic calcium accumulation contributes to muscle degeneration in Duchenne muscular dystrophy (DMD). Sarco/endoplasmic reticulum calcium ATPase (SERCA) is a sarcoplasmic reticulum (SR) calcium pump that actively transports calcium from the cytosol into the SR. We previously showed that adeno-associated virus (AAV)-mediated SERCA2a therapy reduced cytosolic calcium overload and improved muscle and heart function in the murine DMD model. Here, we tested whether AAV SERCA2a therapy could ameliorate muscle disease in the canine DMD model. 7.83 × 1013 vector genome particles of the AAV vector were injected into the extensor carpi ulnaris (ECU) muscles of four juvenile affected dogs. Contralateral ECU muscles received excipient. Three months later, we observed widespread transgene expression and significantly increased SERCA2a levels in the AAV-injected muscles. Treatment improved SR calcium uptake, significantly reduced calpain activity, significantly improved contractile kinetics, and significantly enhanced resistance to eccentric contraction-induced force loss. Nonetheless, muscle histology was not improved. To evaluate the safety of AAV SERCA2a therapy, we delivered the vector to the ECU muscle of adult normal dogs. We achieved strong transgene expression without altering muscle histology and function. Our results suggest that AAV SERCA2a therapy has the potential to improve muscle performance in a dystrophic large mammal.

8.
Methods Mol Biol ; 2587: 303-338, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36401036

RESUMEN

Assessing histological changes is essential for characterizing muscle disease progression and for studying the response to therapies in Duchenne muscular dystrophy (DMD), an X-linked progressive muscle-wasting disease caused by the loss of the dystrophin protein. Canine models are by far the best-characterized large animal models for DMD. In this chapter, we describe methods for muscle tissue collection and storage, hematoxylin and eosin staining for studying general muscle morphology, and special staining protocols for evaluating fibrosis, calcification, and neuronal nitric oxide synthase (nNOS) activity. We also provide immunofluorescence staining protocols that are often used to characterize the expression and localization of dystrophin and components of the dystrophin-associated glycoprotein complex. Lastly, we presented immunohistochemical staining protocols that we use to assess muscle inflammation and immune responses.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Perros , Animales , Distrofina/genética , Distrofina/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Terapia Genética , Músculos/metabolismo
9.
Methods Mol Biol ; 2587: 67-103, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36401025

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal muscle disease caused by dystrophin deficiency. Patients gradually lose motor function, become wheelchair-bound, and die from respiratory and/or cardiac muscle failure. Dystrophin-null dogs have been used as a large animal model for DMD since 1988 and are considered an excellent bridge between rodent models and human patients. While numerous protocols have been published for studying muscle and heart physiology in mice, few such protocols exist for studying skeletal muscle contractility, heart function, and whole-body activity in dogs. Over the last 20 years, we have developed and adapted an array of assays to evaluate whole-body movement, gait, single muscle force, whole limb torque, cardiac electrophysiology, and hemodynamic function in normal and dystrophic dogs. In this chapter, we present detailed working protocols for these assays and lessons we learned during the development and use of these protocols.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Humanos , Perros , Animales , Ratones , Corazón , Músculo Esquelético , Miocardio
10.
Methods Mol Biol ; 2587: 255-301, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36401035

RESUMEN

Mutations in the dystrophin gene result in Duchenne muscular dystrophy (DMD), a progressive muscle-wasting disease. Adeno-associated virus (AAV) mediated gene replacement, and CRISPR/Cas9-mediated genome editing hold the potential to treat DMD. Molecular and biochemical analyses are essential to determine gene transfer efficiency and therapeutic efficacy. In this chapter, we present a series of methods routinely used in our laboratory to extract and quantify DNA, RNA, and protein in gene therapy studies performed in the canine DMD model.


Asunto(s)
Distrofia Muscular de Duchenne , Animales , Perros , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Sistemas CRISPR-Cas/genética , Terapia Genética/métodos , Edición Génica/métodos , Dependovirus/genética
11.
Methods Mol Biol ; 2587: 339-352, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36401037

RESUMEN

Magnetic resonance imaging (MRI) is a well-established and widely used technique to characterize and quantify skeletal and cardiac muscle changes in Duchenne muscular dystrophy (DMD). Recently, MRI has been explored to study disease progression and response to gene therapy in the canine DMD model. Using traditional sequences, delayed gadolinium enhancement, novel sequences, and spectroscopy, investigators have begun to (i) establish the baseline MRI characteristics of the muscles in normal and affected dogs and (ii) evaluate gene therapy outcomes in treated dogs. As a noninvasive assay, MRI offers an excellent opportunity to study longitudinal muscle changes in long-term gene therapy studies in the canine model. In this chapter, we outline the MRI method used to study DMD in the canine model.


Asunto(s)
Distrofia Muscular de Duchenne , Perros , Animales , Distrofia Muscular de Duchenne/diagnóstico por imagen , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Medios de Contraste , Músculo Esquelético/patología , Gadolinio , Imagen por Resonancia Magnética/métodos , Terapia Genética
12.
Methods Mol Biol ; 2587: 353-375, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36401038

RESUMEN

The immune response is a primary hurdle in the development of gene therapy for neuromuscular diseases. Both innate and adaptive immune responses have been observed in human trials. The canine model is an excellent platform to understand immunological consequences of gene therapy. Over the last several decades, we have conducted gene replacement and gene repair therapies in the canine model of Duchenne muscular dystrophy (DMD) using adeno-associated virus (AAV)-mediated expression of the highly abbreviated micro-dystrophin gene, the larger mini-dystrophin gene, and the Cas9-based CRISPR genome editing machinery. We have evaluated the innate, humoral, and cellular immune responses to the AAV vector and the transgene product. In this chapter, we share our experience in collecting and processing of the dog blood samples for immunological assays, and our protocols for quantitative evaluation of cytokines and chemokines, antibodies, and T-cell responses.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Humanos , Perros , Animales , Distrofina/genética , Distrofina/metabolismo , Vectores Genéticos/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Inmunidad Humoral
13.
Muscle Nerve ; 46(6): 948-50, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23225385

RESUMEN

INTRODUCTION: The goal of this study was to determine whether a minimal level of dystrophin expression improves the passive mechanical properties of skeletal muscle in the murine Duchenne muscular dystrophy model. METHODS: We compared the elastic and viscous properties of the extensor digitorum longus muscle (EDL) in mdx3cv and mdx4cv mice at 6, 14, and 20 months of age. Both strains are on the C57Bl/6 background, and both lose the full-length dystrophin protein. Interestingly, mdx3cv mice express a near full-length dystrophin at ≈ 5% of the normal level. RESULTS: We found that the stress-strain profile and the stress relaxation rate of the EDL in mdx3cv mice were partially preserved in all age groups compared with age-matched mdx4cv mice. CONCLUSION: Our results suggest that a low level of dystrophin expression may treat muscle stiffness in Duchenne muscular dystrophy.


Asunto(s)
Distrofina/deficiencia , Regulación de la Expresión Génica , Contracción Muscular/fisiología , Distrofias Musculares , Factores de Edad , Animales , Modelos Animales de Enfermedad , Distrofina/biosíntesis , Hidroxiprolina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/fisiopatología , Distrofia Muscular Animal , Mutación , Estrés Mecánico
14.
Muscle Nerve ; 45(2): 250-6, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22246882

RESUMEN

INTRODUCTION: Duchenne muscular dystrophy (DMD) is a severe, muscle-wasting disease caused by mutations in the dystrophin gene. The mdx mouse is the first and perhaps the most commonly used animal model for study of DMD. Both male and female mdx mice are used. However, it is not completely clear whether gender influences contraction and the passive mechanical properties of mdx skeletal muscle. METHODS: We compared isometric tetanic forces and passive forces of the extensor digitorum longus muscle between male and female mdx mice. RESULTS: At age 6 months, female mdx mice showed better-preserved specific tetanic force. Interestingly, at 20 months of age, female mdx muscle appeared stiffer. CONCLUSIONS: Our results suggest that gender may profoundly influence physiological measurement outcomes in mdx mice.


Asunto(s)
Contracción Muscular/fisiología , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/fisiopatología , Caracteres Sexuales , Animales , Peso Corporal/genética , Femenino , Masculino , Ratones , Ratones Endogámicos mdx , Estrés Mecánico
15.
J Pathol ; 223(1): 88-98, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21125668

RESUMEN

The mechanism of force reduction is not completely understood in Duchenne muscular dystrophy (DMD), a dystrophin-deficient lethal disease. Nitric oxide regulates muscle force. Interestingly, neuronal nitric oxide synthase µ (nNOSµ), a major source of muscle nitric oxide, is lost from the sarcolemma in DMD muscle. We hypothesize that nNOSµ delocalization contributes to force reduction in DMD. To test this hypothesis, we generated dystrophin/nNOSµ double knockout mice. Genetic elimination of nNOSµ significantly enhanced force in dystrophin-null mice. Pharmacological inhibition of nNOS yielded similar results. To further test our hypothesis, we studied δ-sarcoglycan-null mice, a model of limb-girdle muscular dystrophy. These mice had minimal sarcolemmal nNOSµ delocalization and muscle force was less compromised. Annihilation of nNOSµ did not improve their force either. To determine whether nNOSµ delocalization itself inhibited force, we corrected muscle disease in dystrophin-null mice with micro-dystrophins that either restored or did not restore sarcolemmal nNOSµ. Similar muscle force was obtained irrespective of nNOSµ localization. Additional studies suggest that nNOSµ delocalization selectively inhibits muscle force in dystrophin-null mice via nitrosative stress. In summary, we have demonstrated for the first time that nitrosative stress elicited by nNOSµ delocalization is an important mechanism underlying force loss in DMD.


Asunto(s)
Distrofina/deficiencia , Contracción Muscular/fisiología , Distrofia Muscular Animal/metabolismo , Óxido Nítrico Sintasa de Tipo I/deficiencia , Animales , Masculino , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/fisiopatología , Distrofia Muscular de Cinturas/metabolismo , Distrofia Muscular de Cinturas/fisiopatología , Distrofia Muscular Animal/fisiopatología , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatología , Óxido Nítrico Sintasa de Tipo I/fisiología , Estrés Oxidativo/fisiología , Sarcoglicanos/deficiencia , Sarcolema/enzimología
16.
Sci Rep ; 12(1): 4788, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35314731

RESUMEN

This study aims to develop a 4-limb canine gait analysis system using wireless inertial measurement units (IMUs). 3D printed sensor holders were designed to ensure quick and consistent sensor mounting. Signal analysis algorithms were developed to automatically determine the timing of swing start and end in a stride. To evaluate the accuracy of the new system, a synchronized study was conducted in which stride parameters in four dogs were measured simultaneously using the 4-limb IMU system and a pressure-sensor based walkway gait system. The results showed that stride parameters measured in both systems were highly correlated. Bland-Altman analyses revealed a nominal mean measurement bias between the two systems in both forelimbs and hindlimbs. Overall, the disagreement between the two systems was less than 10% of the mean value in over 92% of the data points acquired from forelimbs. The same performance was observed in hindlimbs except for one parameter due to small mean values. We demonstrated that this 4-limb system could successfully visualize the overall gait types and identify rapid gait changes in dogs. This method provides an effective, low-cost tool for gait studies in veterinary applications or in translational studies using dog models of neuromuscular diseases.


Asunto(s)
Miembro Anterior , Marcha , Algoritmos , Animales , Perros , Extremidades , Miembro Posterior
17.
Hum Gene Ther ; 33(9-10): 518-528, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35350865

RESUMEN

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.


Asunto(s)
Distrofina , Distrofia Muscular de Duchenne , Animales , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Dependovirus/metabolismo , Distrofina/genética , Distrofina/metabolismo , Edición Génica/métodos , Terapia Genética/métodos , Ratones , Ratones Endogámicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo
18.
Am J Physiol Heart Circ Physiol ; 301(3): H773-83, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21705671

RESUMEN

To study Ca(2+) signaling in the endothelium of murine feed arteries, we determined the in vitro stability of endothelial cell (EC) tubes freshly isolated from abdominal muscle feed arteries of male and female C57BL/6 mice (5-9 mo, 25-35 g). We tested the hypothesis that intracellular Ca(2+) concentration ([Ca(2+)](i)) responses to muscarinic receptor activation would increase with temperature. Intact EC tubes (length: 1-2 mm, width: 65-80 µm) were isolated using gentle enzymatic digestion with trituration to remove smooth muscle cells. A freshly isolated EC tube was secured in a chamber and superfused at 24 (room temperature), 32, or 37°C. Using fura-2 dye, [Ca(2+)](i) was monitored (ratio of fluorescence at 340- to 380-nm wavelength) at rest and in response to bolus doses of ACh (20 nmol to 200 µmol). The morphological integrity of EC tubes was preserved at 24 and 32°C. Based on the Ca(2+) K(d) values we determined for fura-2 (174 nM at 24°C and 146 nM at 32°C), resting [Ca(2+)](i) remained stable for 180 min at both 24 and 32°C (27 ± 4 and 34 ± 2 nM, respectively), with peak responses to ACh (20 µmol) increasing from ∼220 nM at 24°C to ∼500 nM at 32°C (P < 0.05). There was no difference in responses to ACh between EC tubes from male versus female mice. When EC tubes were maintained at 37°C (typical in vivo temperature), resting [Ca(2+)](i) increased by ∼30% within 15 min, and gaps formed between individual ECs as they retracted and extruded dye, precluding further study. We conclude that EC tubes enable Ca(2+) signaling to be evaluated in the freshly isolated endothelium of murine feed arteries. While Ca(2+) responses are enhanced by approximately twofold at 32 versus 24°C, the instability of EC tubes at 37°C precludes their study at typical body temperature.


Asunto(s)
Músculos Abdominales/irrigación sanguínea , Señalización del Calcio , Forma de la Célula , Células Endoteliales/metabolismo , Temperatura , Acetilcolina/farmacología , Análisis de Varianza , Animales , Arterias/metabolismo , Señalización del Calcio/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Femenino , Fluorometría , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Agonistas Muscarínicos/farmacología
19.
Muscle Nerve ; 43(2): 283-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21254096

RESUMEN

mdx, mdx3cv, and mdx4cv mice are among the most commonly used models for the study of Duchenne muscular dystrophy. Their disease is caused by point mutations in the dystrophin gene. Despite widespread use of these models, genotyping has not always been straightforward. Current methods require multiple polymerase chain reactions (PCRs), post-PCR manipulations, and/or special equipment/reagents. Herein we report a simple, robust PCR genotyping method based on primer competition. This approach could also be applied in genotyping other point-mutation models.


Asunto(s)
Cartilla de ADN/genética , Distrofina/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatología , Reacción en Cadena de la Polimerasa , Animales , Modelos Animales de Enfermedad , Genotipo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Mutación Puntual/genética
20.
Dis Model Mech ; 14(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34704592

RESUMEN

Aged dystrophin-null canines are excellent models for studying experimental therapies for Duchenne muscular dystrophy, a lethal muscle disease caused by dystrophin deficiency. To establish the baseline, we studied the extensor carpi ulnaris (ECU) muscle in 15 terminal age (3-year-old) male affected dogs and 15 age/sex-matched normal dogs. Affected dogs showed histological and anatomical hallmarks of dystrophy, including muscle inflammation and fibrosis, myofiber size variation and centralized myonuclei, as well as a significant reduction of muscle weight, muscle-to-body weight ratio and muscle cross-sectional area. To rigorously characterize the contractile properties of the ECU muscle, we developed a novel in situ assay. Twitch and tetanic force, contraction and relaxation rate, and resistance to eccentric contraction-induced force loss were significantly decreased in affected dogs. Intriguingly, the time-to-peak tension and half-relaxation time were significantly shortened in affected dogs. Contractile kinetics predicted an unforeseen slow-to-fast myofiber-type switch, which we confirmed at the protein and transcript level. Our study establishes a foundation for studying long-term and late-stage therapeutic interventions in dystrophic canines. The unexpected myofiber-type switch highlights the complexity of muscle remodeling in dystrophic large mammals. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Anciano , Animales , Modelos Animales de Enfermedad , Perros , Distrofina/metabolismo , Humanos , Masculino , Mamíferos , Contracción Muscular , Músculo Esquelético/patología , Distrofia Muscular de Duchenne/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA