Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Hum Mol Genet ; 28(3): 396-406, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30281092

RESUMO

Duchenne muscular dystrophy (DMD) is caused by loss of dystrophin protein, leading to progressive muscle weakness and premature death due to respiratory and/or cardiac complications. Cardiac involvement is characterized by progressive dilated cardiomyopathy, decreased fractional shortening and metabolic dysfunction involving reduced metabolism of fatty acids-the major cardiac metabolic substrate. Several mouse models have been developed to study molecular and pathological consequences of dystrophin deficiency, but do not recapitulate all aspects of human disease pathology and exhibit a mild cardiac phenotype. Here we demonstrate that Cmah (cytidine monophosphate-sialic acid hydroxylase)-deficient mdx mice (Cmah-/-;mdx) have an accelerated cardiac phenotype compared to the established mdx model. Cmah-/-;mdx mice display earlier functional deterioration, specifically a reduction in right ventricle (RV) ejection fraction and stroke volume (SV) at 12 weeks of age and decreased left ventricle diastolic volume with subsequent reduced SV compared to mdx mice by 24 weeks. They further show earlier elevation of cardiac damage markers for fibrosis (Ctgf), oxidative damage (Nox4) and haemodynamic load (Nppa). Cardiac metabolic substrate requirement was assessed using hyperpolarized magnetic resonance spectroscopy indicating increased in vivo glycolytic flux in Cmah-/-;mdx mice. Early upregulation of mitochondrial genes (Ucp3 and Cpt1) and downregulation of key glycolytic genes (Pdk1, Pdk4, Ppara), also denote disturbed cardiac metabolism and shift towards glucose utilization in Cmah-/-;mdx mice. Moreover, we show long-term treatment with peptide-conjugated exon skipping antisense oligonucleotides (20-week regimen), resulted in 20% cardiac dystrophin protein restoration and significantly improved RV cardiac function. Therefore, Cmah-/-;mdx mice represent an appropriate model for evaluating cardiac benefit of novel DMD therapeutics.


Assuntos
Monofosfato de Citidina/genética , Distrofina/deficiência , Morfolinos/uso terapêutico , Animais , Cardiomiopatia Dilatada/genética , Carnitina O-Palmitoiltransferase/genética , Fator de Crescimento do Tecido Conjuntivo/análise , Monofosfato de Citidina/fisiologia , Modelos Animais de Doenças , Distrofina/genética , Distrofina/metabolismo , Éxons , Terapia Genética/métodos , Coração/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Oxigenases de Função Mista/metabolismo , Distrofia Muscular de Duchenne/genética , Miocárdio/metabolismo , NADPH Oxidase 4/análise , Oligonucleotídeos Antissenso/genética , Peptídeos/genética , Fenótipo , Volume Sistólico , Proteína Desacopladora 3/genética , Função Ventricular Direita
2.
Hum Mol Genet ; 24(15): 4225-37, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25935000

RESUMO

Splice modulation therapy has shown great clinical promise in Duchenne muscular dystrophy, resulting in the production of dystrophin protein. Despite this, the relationship between restoring dystrophin to established dystrophic muscle and its ability to induce clinically relevant changes in muscle function is poorly understood. In order to robustly evaluate functional improvement, we used in situ protocols in the mdx mouse to measure muscle strength and resistance to eccentric contraction-induced damage. Here, we modelled the treatment of muscle with pre-existing dystrophic pathology using antisense oligonucleotides conjugated to a cell-penetrating peptide. We reveal that 15% homogeneous dystrophin expression is sufficient to protect against eccentric contraction-induced injury. In addition, we demonstrate a >40% increase in specific isometric force following repeated administrations. Strikingly, we show that changes in muscle strength are proportional to dystrophin expression levels. These data define the dystrophin restoration levels required to slow down or prevent disease progression and improve overall muscle function once a dystrophic environment has been established in the mdx mouse model.


Assuntos
Peptídeos Penetradores de Células/genética , Distrofina/biossíntese , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/genética , Animais , Peptídeos Penetradores de Células/administração & dosagem , Modelos Animais de Doenças , Distrofina/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/patologia , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/administração & dosagem
3.
Hum Mol Genet ; 23(7): 1842-55, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24234655

RESUMO

Mutations in fukutin-related protein (FKRP) underlie a group of muscular dystrophies associated with the hypoglycosylation of α-dystroglycan (α-DG), a proportion of which show central nervous system involvement. Our original FKRP knock-down mouse (FKRP(KD)) replicated many of the characteristics seen in patients at the severe end of the dystroglycanopathy spectrum but died perinatally precluding its full phenotyping and use in testing potential therapies. We have now overcome this by crossing FKRP(KD) mice with those expressing Cre recombinase under the Sox1 promoter. Owing to our original targeting strategy, this has resulted in the restoration of Fkrp levels in the central nervous system but not the muscle, thereby generating a new model (FKRP(MD)) which develops a progressive muscular dystrophy resembling what is observed in limb girdle muscular dystrophy. Like-acetylglucosaminyltransferase (LARGE) is a bifunctional glycosyltransferase previously shown to hyperglycosylate α-DG. To investigate the therapeutic potential of LARGE up-regulation, we have now crossed the FKRP(MD) line with one overexpressing LARGE and show that, contrary to expectation, this results in a worsening of the muscle pathology implying that any future strategies based upon LARGE up-regulation require careful management.


Assuntos
Distroglicanas/metabolismo , N-Acetilglucosaminiltransferases/biossíntese , N-Acetilglucosaminiltransferases/genética , Proteínas/genética , Síndrome de Walker-Warburg/genética , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Glicosilação , Laminina/biossíntese , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação , Pentosiltransferases , Transferases , Regulação para Cima , Síndrome de Walker-Warburg/mortalidade
4.
J Neuromuscul Dis ; 8(5): 845-863, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33044191

RESUMO

Duchenne muscular dystrophy is an X-linked, recessive muscular dystrophy in which the absence of the dystrophin protein leads to fibrosis, inflammation and oxidative stress, resulting in loss of muscle tissue. Drug repurposing, i.e. using drugs already approved for other disorders, is attractive as it decreases development time. Recent studies suggested that simvastatin, a cholesterol lowering drug used for cardiovascular diseases, has beneficial effects on several parameters in mdx mice. To validate properly the effectiveness of simvastatin, two independent labs tested the effects of 12-week simvastatin treatment in either young (starting at 4 weeks of age) or adult (starting at 12 weeks of age) mdx mice. In neither study were benefits of simvastatin treatment observed on muscle function, histology or expression of genes involved in fibrosis, regeneration, oxidative stress and autophagy. Unexpectedly, although the treatment protocol was similar, simvastatin plasma levels were found to be much lower than observed in a previous study. In conclusion, in two laboratories, simvastatin did not ameliorate disease pathology in mdx mice, which could either be due to the ineffectiveness of simvastatin itself or due to the low simvastatin plasma levels following oral administration via the food.


Assuntos
Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Sinvastatina/farmacologia , Animais , Modelos Animais de Doenças , Fibrose/fisiopatologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos
5.
Methods Mol Biol ; 423: 421-31, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18370219

RESUMO

Electroporation is a powerful method for gene delivery to dystrophic muscle in the mdx mouse model of Duchenne muscular dystrophy. Successful transfer of reporter and therapeutic plasmids and antisense oligonucleotides has been demonstrated. However, the efficiency falls with increasing plasmid size. Although it is unlikely that the electrotransfer approach will be useful clinically, it is an important experimental tool, particularly in testing potential immune responses to gene transfer in the absence of vector proteins.


Assuntos
Eletroquimioterapia/métodos , Terapia Genética/métodos , Distrofia Muscular Animal/terapia , Animais , DNA Recombinante/administração & dosagem , DNA Recombinante/genética , Distrofina/genética , Distrofina/metabolismo , Expressão Gênica , Genes Reporter , Humanos , Imuno-Histoquímica , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Plasmídeos/administração & dosagem , Plasmídeos/genética , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
6.
Cell Rep ; 23(3): 899-908, 2018 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-29669293

RESUMO

Generating human skeletal muscle models is instrumental for investigating muscle pathology and therapy. Here, we report the generation of three-dimensional (3D) artificial skeletal muscle tissue from human pluripotent stem cells, including induced pluripotent stem cells (iPSCs) from patients with Duchenne, limb-girdle, and congenital muscular dystrophies. 3D skeletal myogenic differentiation of pluripotent cells was induced within hydrogels under tension to provide myofiber alignment. Artificial muscles recapitulated characteristics of human skeletal muscle tissue and could be implanted into immunodeficient mice. Pathological cellular hallmarks of incurable forms of severe muscular dystrophy could be modeled with high fidelity using this 3D platform. Finally, we show generation of fully human iPSC-derived, complex, multilineage muscle models containing key isogenic cellular constituents of skeletal muscle, including vascular endothelial cells, pericytes, and motor neurons. These results lay the foundation for a human skeletal muscle organoid-like platform for disease modeling, regenerative medicine, and therapy development.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Modelos Biológicos , Engenharia Tecidual , Diferenciação Celular , Linhagem da Célula , Humanos , Hidrogéis/química , Desenvolvimento Muscular , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Alicerces Teciduais/química
7.
Sci Rep ; 5: 11632, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26113184

RESUMO

Duchenne muscular dystrophy (DMD) is caused by absence of the integral structural protein, dystrophin, which renders muscle fibres susceptible to injury and degeneration. This ultimately results in cardiorespiratory dysfunction, which is the predominant cause of death in DMD patients, and highlights the importance of therapeutic targeting of the cardiorespiratory system. While there is some evidence to suggest that restoring dystrophin in the diaphragm improves both respiratory and cardiac function, the role of the diaphragm is not well understood. Here using exon skipping oligonucleotides we predominantly restored dystrophin in the diaphragm and assessed cardiac function by MRI. This approach reduced diaphragmatic pathophysiology and markedly improved diaphragm function but did not improve cardiac function or pathophysiology, with or without exercise. Interestingly, exercise resulted in a reduction of dystrophin protein and exon skipping in the diaphragm. This suggests that treatment regimens may require modification in more active patients. In conclusion, whilst the diaphragm is an important respiratory muscle, it is likely that dystrophin needs to be restored in other tissues, including multiple accessory respiratory muscles, and of course the heart itself for appropriate therapeutic outcomes. This supports the requirement of a body-wide therapy to treat DMD.


Assuntos
Diafragma/fisiopatologia , Modelos Animais de Doenças , Coração/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Sequência de Aminoácidos , Animais , Fator Natriurético Atrial , Western Blotting , Diafragma/diagnóstico por imagem , Diafragma/metabolismo , Distrofina/genética , Distrofina/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Morfolinos/química , Morfolinos/farmacologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/genética , NADPH Oxidase 4 , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Peptídeo Natriurético Tipo C/genética , Peptídeo Natriurético Tipo C/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Condicionamento Físico Animal/fisiologia , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Radiografia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Expert Rev Mol Med ; 4(23): 1-23, 2002 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-14585159

RESUMO

Patients with Duchenne muscular dystrophy (DMD), an X-linked lethal muscle-wasting disease, have abnormal expression of the protein dystrophin within their muscle fibres. In the mdx mouse model of this condition, both germline and neonatal somatic gene transfers of dystrophin cDNAs have demonstrated the potential of gene therapy in treating DMD. However, in many DMD patients, there appears to be no dystrophin expression when muscle biopsies are immunostained or western blots are performed. This raises the possibility that the expression of dystrophin following gene transfer might trigger a destructive immune response against this 'neoantigen'. Immune responses can also be generated against the gene transfer vector used to transfect the dystrophic muscle, and the combined immune response could further damage the already inflamed muscle. These problems are now beginning to be investigated in immunocompetent mdx mice. Although much work remains to be done, there are promising indications that these immune responses might not prove as much of a concern as originally envisaged.


Assuntos
Distrofina/genética , Distrofina/imunologia , Terapia Genética , Distrofia Muscular de Duchenne/terapia , Distrofina/metabolismo , Vetores Genéticos/imunologia , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Mutação
9.
Neuromuscul Disord ; 12 Suppl 1: S11-22, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12206790

RESUMO

There is a pressing need to develop new therapeutic approaches to Duchenne muscular dystrophy, an X-linked fatal disease primarily affecting skeletal and cardiac muscle. Gene therapy is an approach that has attracted much interest since the description of the Duchenne muscular dystrophy gene and its mutations in 1987. Since 1990 numerous reporter and dystrophin gene transfer studies have been conducted on muscles of animals but mostly in mice. Experimental protocols have ranged from germ-line gene transfer (via the production of transgenics) to somatic gene transfer studies using viral or non-viral vectors. But what have we actually learned from such studies that can be applied to patients with Duchenne muscular dystrophy? Various dystrophin, utrophin and integrin recombinant cDNAs have been shown to prevent the development of muscular dystrophy in transgenic dystrophic (mdx) mice. Somatic gene transfer prior to the onset of pathology have been shown to prevent the development of the muscular dystrophy in the mdx mouse but the data is less convincing for the beneficial effects of somatic gene transfer following the establishment of pathology. The time of onset and the course of the disease differ substantially between mouse and man and raise concerns about the applicability of gene therapy in man where the disease manifests in utero and the progression is more severe. The other major concern relates to uncertainty over the efficiency of the different vectors in man, particularly as many patients are likely to have encountered the infectious forms of the viruses that are proposed as vectors.


Assuntos
Distrofina/genética , Técnicas de Transferência de Genes , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Animais , Proteínas do Citoesqueleto/genética , DNA Recombinante , Distrofina/deficiência , Técnicas de Transferência de Genes/efeitos adversos , Técnicas de Transferência de Genes/tendências , Terapia Genética/efeitos adversos , Vetores Genéticos/imunologia , Humanos , Integrinas/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos mdx , Músculos/patologia , Distrofias Musculares/terapia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Utrofina
10.
Neuromuscul Disord ; 13(1): 21-31, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12467729

RESUMO

A lack of effective treatments for Duchenne muscular dystrophy, a fatal X-linked myopathy, has focused attention on the possibility of gene therapy. The aim of the gene therapy approach is the restoration of the dystrophin associated complex of proteins, one member of which is neuronal nitric oxide synthase, an important enzyme in signal transduction. Transgenic mdx mice and plasmid gene transfer of both human and murine recombinant dystrophins was used to assess whether nNOS could be restored to the sarcolemma following dystrophin gene transfer at a variety of levels of expression. Murine revertant fibres and human patients with different dystrophin deletions were used to assess the relationship between exon deletion and loss of neuronal nitric oxide synthase localization to the sarcolemma. We demonstrate that the domain encoded by exons 45-48 is required for localization of neuronal nitric oxide synthase to the sarcolemma. On the basis of these observations we suggest that neuronal nitric oxide synthase is a useful marker for complete restoration of the dystrophin associated complex and should be used as one of the criteria for selecting the recombinant molecule to be used for gene therapy in Duchenne muscular dystrophy.


Assuntos
Distrofina/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Óxido Nítrico Sintase/metabolismo , Adulto , Animais , Biomarcadores/análise , Proteínas de Ligação ao Cálcio , Proteínas do Citoesqueleto/metabolismo , Éxons , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica/métodos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Óxido Nítrico Sintase Tipo I , Fragmentos de Peptídeos/metabolismo , Sarcolema/enzimologia , Utrofina
11.
J Biol Chem ; 278(22): 19956-65, 2003 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-12639970

RESUMO

The 27-kDa heat shock protein (HSP27) has a potent ability to increase cell survival in response to a wide range of cellular challenges. In order to investigate the mode of action of HSP27 in vivo, we have developed transgenic lines, which express human HSP27 at high levels throughout the brain, spinal cord, and other tissues. In view of the particular property of HSP27 compared with other HSPs to protect neurons against apoptosis, we have tested these transgenic lines in a well established in vivo model of neurotoxicity produced by kainic acid, where apoptotic cell death occurs. Our results demonstrate for the first time the marked protective effects of HSP27 overexpression in vivo, which significantly reduces kainate-induced seizure severity and mortality rate (>50%) in two independent lines and markedly reduces neuronal cell death in the CA3 region of hippocampus. This reduced seizure severity in HSP27 transgenic animals was associated with a marked attenuation of caspase 3 induction and apoptotic features. These studies clearly demonstrate that HSP27 has a major neuroprotective effect in the central nervous system in keeping with its properties demonstrated in culture and highlight an early stage in the cell death pathway that is affected by HSP27.


Assuntos
Morte Celular/genética , Proteínas de Choque Térmico , Hipocampo/patologia , Ácido Caínico/toxicidade , Proteínas de Neoplasias/genética , Fármacos Neuroprotetores , Convulsões/prevenção & controle , Animais , Comportamento Animal/efeitos dos fármacos , Caspase 3 , Caspases/biossíntese , Ativação Enzimática , Hipocampo/enzimologia , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares , Convulsões/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA