Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Hum Mol Genet ; 28(13): 2189-2200, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-30990876

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal neuromuscular disorder caused by loss of dystrophin. Several therapeutic modalities are currently in clinical trials but none will achieve maximum functional rescue and full disease correction. Therefore, we explored the potential of combining the benefits of dystrophin with increases of utrophin, an autosomal paralogue of dystrophin. Utrophin and dystrophin can be co-expressed and co-localized at the same muscle membrane. Wild-type (wt) levels of dystrophin are not significantly affected by a moderate increase of utrophin whereas higher levels of utrophin reduce wt dystrophin, suggesting a finite number of actin binding sites at the sarcolemma. Thus, utrophin upregulation strategies may be applied to the more mildly affected Becker patients with lower dystrophin levels. Whereas increased dystrophin in wt animals does not offer functional improvement, overexpression of utrophin in wt mice results in a significant supra-functional benefit over wt. These findings highlight an additive benefit of the combined therapy and potential new unique roles of utrophin. Finally, we show a 30% restoration of wt dystrophin levels, using exon-skipping, together with increased utrophin levels restores dystrophic muscle function to wt levels offering greater therapeutic benefit than either single approach alone. Thus, this combination therapy results in additive functional benefit and paves the way for potential future combinations of dystrophin- and utrophin-based strategies.


Asunto(s)
Distrofina/genética , Distrofia Muscular de Duchenne/terapia , Utrofina/genética , Animales , Distrofina/metabolismo , Exones , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Morfolinos/síntesis química , Morfolinos/uso terapéutico , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Miofibrillas/metabolismo , Sarcolema/metabolismo , Regulación hacia Arriba , Utrofina/metabolismo
2.
Hum Mol Genet ; 28(2): 307-319, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30304405

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. Constitutive utrophin expression, a structural and functional paralogue of dystrophin, can successfully prevent the dystrophic pathology in the dystrophin-deficient mdx mouse model. In dystrophic muscles, utrophin is increased as part of the repair process and localized at the sarcolemma of regenerating myofibers. The presence of developmental myosin such as embryonic myosin (MyHC-emb) and neonatal represents a useful marker of muscle regeneration and a meaningful indicator of muscle damage, which correlates with the clinical severity of milder Becker muscular dystrophy and DMD patients. In the present study, we demonstrate that MyHC-emb is a robust marker of regeneration at different ages and in different skeletal muscles. We also evaluate the correlation between utrophin, dystrophin and MyHC-emb in wild-type (wt) and regenerating dystrophic muscles. Restoration of dystrophin significantly reduced MyHC-emb levels. Similarly, overexpression of utrophin in the transgenic mdx-Fiona mice reduced the number of MyHC-emb positive fibers to wt level, prevented the regenerative process and rescued the muscle function. In contrast, the absence of utrophin in the dystrophin-deficient double-knockout mice resulted in a higher MyHC-emb content and in a more severe dystrophic pathophysiology than in mdx mice. These data illustrate the importance of monitoring utrophin and MyHC-emb levels in the preclinical evaluation of therapies and provide translational support for the use of developmental myosin as a disease biomarker in DMD clinical trials.


Asunto(s)
Distrofina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Miosinas/metabolismo , Regeneración , Utrofina/metabolismo , Animales , Biomarcadores/metabolismo , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Embrión de Mamíferos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/embriología , Músculo Esquelético/fisiología , Distrofia Muscular Animal , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/embriología , Distrofia Muscular de Duchenne/patología , Sarcolema/metabolismo
3.
Tetrahedron ; 76(2): 130819, 2020 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-32713969

RESUMEN

Following on from ezutromid, the first-in-class benzoxazole utrophin modulator that progressed to Phase 2 clinical trials for the treatment of Duchenne muscular dystrophy, a new chemotype was designed to optimise its physicochemical and ADME profile. Herein we report the synthesis of SMT022357, a second generation utrophin modulator preclinical candidate, and an asymmetric synthesis of its constituent enantiomers. The pharmacological properties of both enantiomers were evaluated in vitro and in vivo. No significant difference in the activity or efficacy was observed between the two enantiomers; activity was found to be comparable to the racemic mixture.

4.
Annu Rev Genomics Hum Genet ; 16: 281-308, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26048046

RESUMEN

Current molecular genomic approaches to human genetic disorders have led to an explosion in the identification of the genes and their encoded proteins responsible for these disorders. The identification of the gene altered by mutations in Duchenne and Becker muscular dystrophy was one of the earliest examples of this paradigm. The nearly 30 years of research partly outlined here exemplifies the road that similar current gene discovery protocols will be expected to travel, albeit much more rapidly owing to improved diagnosis of genetic disorders and an understanding of the spectrum of mutations thought to cause them. The identification of the protein dystrophin has led to a new understanding of the muscle cell membrane and the proteins involved in membrane stability, as well as new candidate genes for additional forms of muscular dystrophy. Animal models identified with naturally occurring mutations and developed by genetic manipulation have furthered the understanding of disease progression and underlying pathology. The biochemistry and molecular analysis of patient samples have led to the different dystrophin-dependent and -independent therapies that are currently close to or in human clinical trials. The lessons learned from decades of research on dystrophin have benefited the field of human genetics.


Asunto(s)
Distrofina/metabolismo , Distrofias Musculares/fisiopatología , Distrofias Musculares/terapia , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Distrofina/genética , Terapia Genética/métodos , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Hidrolasas Diéster Fosfóricas/metabolismo , Esteroides/uso terapéutico , Utrofina/genética , Utrofina/metabolismo
5.
Am J Pathol ; 187(3): 505-516, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28056338

RESUMEN

Collagen IV is a major component of basement membranes (BMs). The α1(IV) chain, encoded by the COL4A1 gene, is expressed ubiquitously and associates with the α2(IV) chain to form the α1α1α2(IV) heterotrimer. Several COL4A1 mutations affecting a conformational domain containing integrin-binding sites are responsible for the systemic syndrome of hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC). To analyze the pathophysiology of HANAC, Col4a1 mutant mice bearing the p.Gly498Val mutation were generated. Analysis of the skeletal muscles of Col4a1G498V mutant animals showed morphologic characteristics of a muscular dystrophy phenotype with myofiber atrophy, centronucleation, focal inflammatory infiltrates, and fibrosis. Abnormal ultrastructural aspects of muscle BMs was associated with reduced extracellular secretion of the mutant α1α1α2(IV) trimer. In addition to muscular dystrophic features, endothelial cell defects of the muscle capillaries were observed, with intracytoplasmic accumulation of the mutant α1α1α2(IV) molecules, endoplasmic reticulum cisternae dilation, and up-regulation of endoplasmic reticulum stress markers. Induction of the unfolded protein response in Col4a1 mutant muscle tissue resulted in an excess of apoptosis in endothelial cells. HANAC mutant animals also presented with a muscular functional impairment and increased serum creatine kinase levels reflecting altered muscle fiber sarcolemma. This extensive description of the muscular phenotype of the Col4a1 HANAC murine model suggests a potential contribution of primary endothelial cell defects, together with muscle BM alterations, to the development of COL4A1-related myopathy.


Asunto(s)
Vasos Sanguíneos/anomalías , Colágeno Tipo IV/genética , Calambre Muscular/genética , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/patología , Mutación/genética , Enfermedad de Raynaud/genética , Animales , Apoptosis , Vasos Sanguíneos/patología , Peso Corporal , Creatina Quinasa/sangre , Distrofina/metabolismo , Estrés del Retículo Endoplásmico , Células Endoteliales/patología , Células Endoteliales/ultraestructura , Matriz Extracelular/metabolismo , Integrina beta1/metabolismo , Ratones , Ratones Mutantes , Músculo Esquelético/ultraestructura , Tamaño de los Órganos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
6.
Hum Mol Genet ; 24(15): 4212-24, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25935002

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.


Asunto(s)
Distrofina/biosíntesis , Fibras Musculares Esqueléticas/efectos de los fármacos , Distrofia Muscular de Duchenne/tratamiento farmacológico , Utrofina/biosíntesis , Animales , Distrofina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos mdx , Contracción Muscular/efectos de los fármacos , Contracción Muscular/genética , Fibras Musculares Esqueléticas/patología , Músculos/efectos de los fármacos , Músculos/patología , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Sarcolema/efectos de los fármacos , Sarcolema/genética , Utrofina/genética
7.
Exp Physiol ; 100(12): 1458-67, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26140505

RESUMEN

NEW FINDINGS: What is the topic of this review? This review highlights recent progress in genetically based therapies targeting the primary defect of Duchenne muscular dystrophy. What advances does it highlight? Over the last two decades, considerable progress has been made in understanding the mechanisms underlying Duchenne muscular dystrophy, leading to the development of genetic therapies. These include manipulation of the expression of the gene or related genes, the splicing of the gene and its translation, and replacement of the gene using viral approaches. Duchenne muscular dystrophy is a lethal X-linked disorder caused by mutations in the dystrophin gene. In the absence of the dystrophin protein, the link between the cytoskeleton and extracellular matrix is destroyed, and this severely compromises the strength, flexibility and stability of muscle fibres. The devastating consequence is progressive muscle wasting and premature death in Duchenne muscular dystrophy patients. There is currently no cure, and despite exhaustive palliative care, patients are restricted to a wheelchair by the age of 12 years and usually succumb to cardiac or respiratory complications in their late 20s. This review provides an update on the current genetically based therapies and clinical trials that target or compensate for the primary defect of this disease. These include dystrophin gene-replacement strategies, genetic modification techniques to restore dystrophin expression, and modulation of the dystrophin homologue, utrophin, as a surrogate to re-establish muscle function.


Asunto(s)
Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Animales , Ensayos Clínicos como Asunto , Distrofina/genética , Terapia Genética/métodos , Humanos , Músculo Esquelético/metabolismo , Utrofina/genética
9.
Cells ; 13(10)2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38786024

RESUMEN

In recent years, clustered regularly interspaced short palindromic repeats (CRISPRs) and CRISPR-associated (Cas) protein have emerged as a revolutionary gene editing tool to treat inherited disorders affecting different organ systems, such as blood and muscles. Both hematological and neuromuscular genetic disorders benefit from genome editing approaches but face different challenges in their clinical translation. The ability of CRISPR/Cas9 technologies to modify hematopoietic stem cells ex vivo has greatly accelerated the development of genetic therapies for blood disorders. In the last decade, many clinical trials were initiated and are now delivering encouraging results. The recent FDA approval of Casgevy, the first CRISPR/Cas9-based drug for severe sickle cell disease and transfusion-dependent ß-thalassemia, represents a significant milestone in the field and highlights the great potential of this technology. Similar preclinical efforts are currently expanding CRISPR therapies to other hematologic disorders such as primary immunodeficiencies. In the neuromuscular field, the versatility of CRISPR/Cas9 has been instrumental for the generation of new cellular and animal models of Duchenne muscular dystrophy (DMD), offering innovative platforms to speed up preclinical development of therapeutic solutions. Several corrective interventions have been proposed to genetically restore dystrophin production using the CRISPR toolbox and have demonstrated promising results in different DMD animal models. Although these advances represent a significant step forward to the clinical translation of CRISPR/Cas9 therapies to DMD, there are still many hurdles to overcome, such as in vivo delivery methods associated with high viral vector doses, together with safety and immunological concerns. Collectively, the results obtained in the hematological and neuromuscular fields emphasize the transformative impact of CRISPR/Cas9 for patients affected by these debilitating conditions. As each field suffers from different and specific challenges, the clinical translation of CRISPR therapies may progress differentially depending on the genetic disorder. Ongoing investigations and clinical trials will address risks and limitations of these therapies, including long-term efficacy, potential genotoxicity, and adverse immune reactions. This review provides insights into the diverse applications of CRISPR-based technologies in both preclinical and clinical settings for monogenic blood disorders and muscular dystrophy and compare advances in both fields while highlighting current trends, difficulties, and challenges to overcome.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Terapia Genética , Humanos , Terapia Genética/métodos , Sistemas CRISPR-Cas/genética , Animales , Edición Génica/métodos , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/genética , Ensayos Clínicos como Asunto , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética
10.
Med ; 4(4): 220-222, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-37060897

RESUMEN

This month in Med, the description of an unusually severely affected DMD patient suffering from a large deletion in the dystrophin gene confirms that absence of utrophin worsens the dystrophy and supports the concept that utrophin upregulation ameliorates the pathology. This study may guide the development of dystrophin-based gene therapies.


Asunto(s)
Distrofia Muscular de Duchenne , Humanos , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofina/genética , Distrofina/metabolismo , Utrofina/genética , Utrofina/metabolismo , Regulación hacia Arriba , Gravedad del Paciente
11.
J Med Chem ; 63(14): 7880-7891, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32551645

RESUMEN

Utrophin modulation is a promising therapeutic strategy for Duchenne muscular dystrophy (DMD), which should be applicable to all patient populations. Following on from ezutromid, the first-generation utrophin modulator, we describe the development of a second generation of utrophin modulators, based on the bioisosteric replacement of the sulfone group with a phosphinate ester and substitution of the metabolically labile naphthalene with a haloaryl substituent. The improved physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties, further reflected in the enhanced pharmacokinetic profile of the most advanced compounds, 30 and 27, led to significantly better in vivo exposure compared to ezutromid and alleviation of the dystrophic phenotype in mdx mice. While 30 was found to have dose-limiting hepatotoxicity, 27 and its enantiomers exhibited limited off-target effects, resulting in a safe profile and highlighting their potential utility as next-generation utrophin modulators suitable for progression toward a future DMD therapy.


Asunto(s)
Benzoxazoles/uso terapéutico , Distrofia Muscular de Duchenne/tratamiento farmacológico , Utrofina/metabolismo , Animales , Benzoxazoles/síntesis química , Benzoxazoles/farmacocinética , Benzoxazoles/toxicidad , Escherichia coli/efectos de los fármacos , Ratones Endogámicos mdx , Estructura Molecular , Distrofia Muscular de Duchenne/metabolismo , Pruebas de Mutagenicidad , Ratas , Salmonella typhimurium/efectos de los fármacos , Estereoisomerismo , Relación Estructura-Actividad , Regulación hacia Arriba/efectos de los fármacos
12.
Neural Regen Res ; 14(8): 1317-1320, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30964048

RESUMEN

Skeletal muscle has an extraordinary capacity to regenerate after injury and trauma. The muscle repair mechanism is a complex process orchestrated by multiple steps. In neuromuscular disorders such as Duchenne muscular dystrophy (DMD), the pathological consequences of the lack of dystrophin and the loss of the dystrophin-associated protein complex are dramatic, with a progressive cascade of events, such as continual influx of inflammation, repeated cycles of degeneration and impaired regeneration. Thus, muscle regeneration is a hallmark of the disease and careful monitoring of regenerative processes with robust markers should provide useful information to the field. Since decades, several indices of regeneration such as centronucleation and fibre size have been commonly used. In the present review, we discuss the impaired regenerative process in DMD, the common and new indices of regeneration and their associated methodologies. We notably highlight the regenerative marker embryonic myosin as a robust indicator of muscle regeneration. We also describe new quantitative methodologies offering the possibility of using a panel of translational regenerative biomarkers to obtain a more complete view of the regeneration processes. Upregulation of utrophin, an autosomal and functional paralogue of dystrophin, is one of the most promising therapeutic strategies as it targets the primary cause of the disease and is applicable to all DMD patients regardless their genetic defects. As utrophin is a regeneration associated protein increased in dystrophic muscle, we discuss the correlation of utrophin levels after drug treatment with regeneration markers. The recent advances in technologies and complementary markers of muscle regeneration described in this review, provide an unprecedented opportunity to develop more robust utrophin DMD based strategies for all DMD patients.

13.
Mol Ther Methods Clin Dev ; 11: 92-105, 2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30417024

RESUMEN

Duchenne muscular dystrophy (DMD) is an X-linked muscle-wasting disease caused by mutations in the dystrophin gene. DMD boys are wheelchair-bound around 12 years and generally survive into their twenties. There is currently no effective treatment except palliative care, although personalized treatments such as exon skipping, stop codon read-through, and viral-based gene therapies are making progress. Patients present with skeletal muscle pathology, but most also show cardiomyopathy by the age of 10. A systemic therapeutic approach is needed that treats the heart and skeletal muscle defects in all patients. The dystrophin-related protein utrophin has been shown to compensate for the lack of dystrophin in the mildly affected BL10/mdx mouse. The purpose of this investigation was to demonstrate that AAV9-mediated micro-utrophin transgene delivery can not only functionally replace dystrophin in the heart, but also attenuate the skeletal muscle phenotype in severely affected D2/mdx mice. The data presented here show that utrophin can indeed alleviate the pathology in skeletal and cardiac muscle in D2/mdx mice. These results endorse the view that utrophin modulation has the potential to increase the quality life of all DMD patients whatever their mutation.

14.
Curr Opin Pharmacol ; 34: 36-48, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28486179

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of dystrophin, essential for muscle fibre integrity. Despite extensive pre-clinical studies, development of an effective treatment has proved challenging. More recently, significant progress has been made with the first drug approval using a genetic approach and the application of pharmacological agents which slow the progression of the disease. Drug development for DMD has mainly used two strategies: (1) the restoration of dystrophin expression or the expression of the compensatory utrophin protein as an efficient surrogate, and (2) the mitigation of secondary downstream pathological mechanisms. This review details current most promising pharmacological approaches and clinical trials aiming to tackle the pathogenesis of this multifaceted disorder.


Asunto(s)
Distrofia Muscular de Duchenne/tratamiento farmacológico , Animales , Distrofina/deficiencia , Complejo de Proteínas Asociado a la Distrofina/metabolismo , Humanos , Distrofia Muscular de Duchenne/metabolismo
15.
Sci Rep ; 7: 43697, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28252048

RESUMEN

Despite promising therapeutic avenues, there is currently no effective treatment for Duchenne muscular dystrophy (DMD), a lethal monogenic disorder caused by the loss of the large cytoskeletal protein, dystrophin. A highly promising approach to therapy, applicable to all DMD patients irrespective to their genetic defect, is to modulate utrophin, a functional paralogue of dystrophin, able to compensate for the primary defects of DMD restoring sarcolemmal stability. One of the major difficulties in assessing the effectiveness of therapeutic strategies is to define appropriate outcome measures. In the present study, we utilised an aptamer based proteomics approach to profile 1,310 proteins in plasma of wild-type, mdx and Fiona (mdx overexpressing utrophin) mice. Comparison of the C57 and mdx sera revealed 83 proteins with statistically significant >2 fold changes in dystrophic serum abundance. A large majority of previously described biomarkers (ANP32B, THBS4, CAMK2A/B/D, CYCS, CAPNI) were normalised towards wild-type levels in Fiona animals. This work also identified potential mdx markers specific to increased utrophin (DUS3, TPI1) and highlights novel mdx biomarkers (GITR, MYBPC1, HSP60, SIRT2, SMAD3, CNTN1). We define a panel of putative protein mdx biomarkers to evaluate utrophin based strategies which may help to accelerate their translation to the clinic.


Asunto(s)
Biomarcadores , Proteínas Sanguíneas , Utrofina/sangre , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Ratones Transgénicos , Distrofia Muscular Animal , Distrofia Muscular de Duchenne/sangre , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Proteoma , Proteómica/métodos , Investigación Biomédica Traslacional , Utrofina/uso terapéutico
16.
Skelet Muscle ; 7(1): 22, 2017 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-29065908

RESUMEN

BACKGROUND: Duchenne muscular dystrophy (DMD) is a lethal X-linked muscle wasting disorder caused by the absence of dystrophin, a large cytoskeletal muscle protein. Increasing the levels of the dystrophin-related-protein utrophin is a highly promising therapy for DMD and has been shown to improve pathology in dystrophin-deficient mice. One contributing factor to muscle wasting in DMD is mitochondrial pathology that contributes to oxidative stress and propagates muscle damage. The purpose of this study was to assess whether utrophin could attenuate mitochondria pathology and oxidative stress. METHODS: Skeletal muscles from wildtype C57BL/10, dystrophin-deficient mdx, dystrophin/utrophin double knockout (dko) and dystrophin-deficient mdx/utrophin over-expressing mdx-Fiona transgenic mice were assessed for markers of mitochondrial damage. RESULTS: Using transmission electron microscopy, we show that high levels of utrophin ameliorate the aberrant structure and localisation of mitochondria in mdx mice whereas absence of utrophin worsened these features in dko mice. Elevated utrophin also reverts markers of protein oxidation and oxidative stress, elevated in mdx and dko mice, to wildtype levels. These changes were observed independently of a shift in oxidative phenotype. CONCLUSION: These findings show that utrophin levels influence mitochondrial pathology and oxidative stress. While utrophin deficiency worsens the pathology, utrophin over-expression in dystrophic muscle benefits mitochondria and attenuates the downstream pathology associated with aberrant mitochondrial function.


Asunto(s)
Mitocondrias Musculares/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Estrés Oxidativo , Utrofina/genética , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Mitocondrias Musculares/ultraestructura , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Utrofina/metabolismo
17.
Neuromuscul Disord ; 22(12): 1057-68, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22854012

RESUMEN

Decorin is a member of the small leucine-rich proteoglycan family and it is a component of the extracellular matrix. Decorin was previously shown to bind different molecules, including myostatin, in a zinc-dependent manner. Here, we investigated in detail the anti-myostatin activity of decorin and fragments thereof. We show that this protein displays in vitro anti-myostatin activities with an IC(50) of 2.3 × 10(-8)M. After intramuscular injection of decorin in dystrophic mdx and γ-sarcoglycan(-/-) mice, we observed a significant increase of the muscle mass and this effect was maximal 18 days after administration. Further, we show that the myostatin-binding site is located in the N-terminal domain of decorin. In fact, a peptide encompassing the 31-71 sequence retains full myostatin binding capacity and intramuscular injection of the peptide induces muscle hypertrophy. The evaluation of three additional peptides suggests a crucial role of the four cysteines within the conserved CX3CXCX6C motif of class I of the small leucine-rich proteoglycans. Altogether, our results show that the N-terminal domain of decorin is sufficient for the binding to myostatin and they underscore the crucial role for this interaction of zinc and the cysteine cluster.


Asunto(s)
Decorina/farmacología , Enfermedades Musculares/tratamiento farmacológico , Miostatina/antagonistas & inhibidores , Péptidos/farmacología , Zinc/metabolismo , Animales , Decorina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Ratones , Ratones Endogámicos mdx , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Péptidos/metabolismo , Unión Proteica/fisiología , Proteoglicanos/farmacología
18.
Macromol Biosci ; 11(5): 590-4, 2011 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-21337518

RESUMEN

Muscle is an important and attractive target for gene therapy. Recent findings have shown that neutral amphiphilic triblock copolymers with a PEO-PPO-PEO arrangement significantly increase muscle transfection as compared to naked DNA. We were interested in evaluating whether reverse Pluronics (PPO-PEO-PPO) also possess transfection properties. Therefore, we measured the in vitro and in vivo transfection activity of 25R2 and 25R4, two copolymers that differ by their hydrophilic/hydrophobic balance. The results show that 25R2 significantly increases the transfection level in muscle compared to naked DNA. Taken together, this work demonstrates that the reverse Pluronic 25R2 possesses interesting properties for in vivo transfection.


Asunto(s)
ADN/administración & dosificación , Músculo Esquelético/enzimología , Poloxámero/química , Polietilenglicoles/química , Glicoles de Propileno/química , Tensoactivos/química , Transfección/métodos , Animales , Ensayo de Cambio de Movilidad Electroforética , Elementos de Facilitación Genéticos , Femenino , Genes Reporteros , Células HEK293 , Células Hep G2 , Humanos , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA