Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Neuromuscul Dis ; 8(s2): S369-S381, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34569970

RESUMO

BACKGROUND: Phosphorodiamidate morpholino oligomer (PMO)-mediated exon skipping is currently used in clinical development to treat Duchenne muscular dystrophy (DMD), with four exon-skipping drugs achieving regulatory approval. Exon skipping elicits a truncated, but semi-functional dystrophin protein, similar to the truncated dystrophin expressed in patients with Becker Muscular dystrophy (BMD) where the disease phenotype is less severe than DMD. Despite promising results in both dystrophic animal models and DMD boys, restoration of dystrophin by exon skipping is highly variable, leading to contradictory functional outcomes in clinical trials. OBJECTIVE: To develop optimal PMO dosing protocols that result in increased dystrophin and improved outcome measures in preclinical models of DMD. METHODS: Tested effectiveness of multiple chronic, high dose PMO regimens using biochemical, histological, molecular, and imaging techniques in mdx mice. RESULTS: A chronic, monthly regimen of high dose PMO increased dystrophin rescue in mdx mice and improved specific force in the extensor digitorum longus (EDL) muscle. However, monthly high dose PMO administration still results in variable dystrophin expression localized throughout various muscles. CONCLUSIONS: High dose monthly PMO administration restores dystrophin expression and increases muscle force; however, the variability of dystrophin expression at both the inter-and intramuscular level remains. Additional strategies to optimize PMO uptake including increased dosing frequencies or combination treatments with other yet-to-be-defined therapies may be necessary to achieve uniform dystrophin restoration and increases in muscle function.


Assuntos
Distrofina/efeitos dos fármacos , Morfolinos/administração & dosagem , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Animais , Modelos Animais de Doenças , Éxons , Terapia Genética , Masculino , Camundongos , Camundongos Endogâmicos mdx
2.
J Neuromuscul Dis ; 8(4): 469-479, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33523015

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is a rare, X-linked, fatal, degenerative neuromuscular disease caused by DMD gene mutations. A relationship between exon skipping and dystrophin production in exon 51-amenable patients treated with eteplirsen (EXONDYS 51®) is established. Once-weekly eteplirsen significantly increased dystrophin, with slower decline in ambulatory function compared to baseline. Long-term treatment with eteplirsen leads to accumulation of dystrophin over time and observed functional benefits in patients with DMD. OBJECTIVE: Compare long-term ambulatory function in eteplirsen-treated patients versus controls. METHODS: Study 201/202 included 12 eteplirsen-treated patients assessed twice/year for ambulatory function over 4 years. Ambulatory evaluations (6-minute walk test [6MWT], loss of ambulation, and North Star Ambulatory Assessment [NSAA]) were compared with matched controls from Italian Telethon and Leuven registries. RESULTS: At Years 3 and 4, eteplirsen-treated patients demonstrated markedly greater mean 6MWT than controls (difference in change from baseline of 132 m [95%CI (29, 235), p = 0.015] at Year 3 and 159 m [95%CI (66, 253), p = 0.002] at Year 4). At Year 4, a significantly greater proportion of eteplirsen-treated patients were still ambulant versus controls (10/12 vs 3/11; p = 0.020). At Year 3, eteplirsen-treated patients demonstrated milder NSAA decline versus controls (difference in change from baseline of 2.6, 95%CI [-6, 11]), however, the difference was not statistically significant; Year 4 control NSAA data were not available. CONCLUSION: In this retrospective matched control study, eteplirsen treatment resulted in attenuation of ambulatory decline over a 4-year observation period, supporting long-term benefit in patients with DMD.


Assuntos
Morfolinos/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Caminhada , Adolescente , Estudos de Casos e Controles , Criança , Distrofina/efeitos dos fármacos , Éxons , Humanos , Masculino , Mutação , Estudos Retrospectivos , Teste de Caminhada
3.
Int J Exp Pathol ; 102(1): 11-21, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33296126

RESUMO

Dystrophin deficiency makes the sarcolemma fragile and susceptible to degeneration in Duchenne muscular dystrophy. The proteasome is a multimeric protease complex and is central to the regulation of cellular proteins. Previous studies have shown that proteasome inhibition improved pathological changes in mdx mice. Ixazomib is the first oral proteasome inhibitor used as a therapy in multiple myeloma. This study investigated the effects of ixazomib on the dystrophic muscle of mdx mice. MDX mice were treated with ixazomib (7.5 mg/kg/wk by gavage) or 0.2 mL of saline for 12 weeks. The Kondziela test was performed to measure muscle strength. The tibialis anterior (TA) and diaphragm (DIA) muscles were used for morphological analysis, and blood samples were collected for biochemical measurement. We observed maintenance of the muscle strength in the animals treated with ixazomib. Treatment with ixazomib had no toxic effect on the mdx mouse. The morphological analysis showed a reduction in the inflammatory area and fibres with central nuclei in the TA and DIA muscles and an increase in the number of fibres with a diameter of 20 µm2 in the DIA muscle after treatment with ixazomib. There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-ß in the DIA muscle of mdx mice treated with ixazomib. Ixazomib was thus shown to increase the expression of dystrophin and utrophin associated with improved pathological and functional changes in the dystrophic muscles of mdx mice.


Assuntos
Compostos de Boro/farmacologia , Distrofina/efeitos dos fármacos , Glicina/análogos & derivados , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne , Inibidores de Proteases/farmacologia , Animais , Distrofina/metabolismo , Glicina/farmacologia , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Utrofina/efeitos dos fármacos , Utrofina/metabolismo
4.
Ann Clin Transl Neurol ; 7(12): 2393-2408, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33285037

RESUMO

OBJECTIVE: The novel morpholino antisense oligonucleotide viltolarsen targets exon 53 of the dystrophin gene, and could be an effective treatment for patients with Duchenne muscular dystrophy (DMD). We investigated viltolarsen's ability to induce dystrophin expression and examined its safety in DMD patients. METHODS: In this open-label, multicenter, parallel-group, phase 1/2, exploratory study, 16 ambulant and nonambulant males aged 5-12 years with DMD received viltolarsen 40 or 80 mg/kg/week via intravenous infusion for 24 weeks. Primary endpoints were dystrophin expression and exon 53 skipping levels. RESULTS: In western blot analysis, mean changes in dystrophin expression (% normal) from baseline to Weeks 12 and 24 were - 1.21 (P = 0.5136) and 1.46 (P = 0.1636), respectively, in the 40 mg/kg group, and 0.76 (P = 0.2367) and 4.81 (P = 0.0536), respectively, in the 80 mg/kg group. The increase in mean dystrophin level at Weeks 12 and 24 was significant in the 80 mg/kg group (2.78%; P = 0.0364). Patients receiving 80 mg/kg showed a higher mean exon 53 skipping level (42.4%) than those receiving 40 mg/kg (21.8%). All adverse events were judged to be mild or moderate in intensity and none led to study discontinuation. INTERPRETATION: Treatment with viltolarsen 40 or 80 mg/kg elicited an increasing trend in dystrophin expression and exon 53 skipping levels, and was safe and well tolerated. The decline in motor function appeared less marked in patients with higher dystrophin levels; this may warrant further investigation. This study supports the potential clinical benefit of viltolarsen.


Assuntos
Distrofina/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos/farmacologia , Criança , Pré-Escolar , Distrofina/genética , Distrofina/metabolismo , Humanos , Japão , Masculino , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/fisiopatologia , Oligonucleotídeos/administração & dosagem , Oligonucleotídeos/efeitos adversos , Oligonucleotídeos/farmacocinética , Avaliação de Resultados em Cuidados de Saúde
5.
Arq Neuropsiquiatr ; 77(8): 579-589, 2019 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-31508685

RESUMO

In this review, we discuss the therapies used in the treatment of patients with Duchenne muscular dystrophy since the first description of the disease. A short description is given of the various theories based on disease pathogenesis, which give the substrates for the many therapeutic interventions. A brief review of the methods of evaluation used in therapeutic trials is made. Of all the treatments, the only drugs that are still considered able to modify the course of the disease are the corticosteroids (prednisone/prednisolone/deflazacort). Other drugs (coenzyme Q10 and creatine) have had a little effect in a few functions without adverse reactions. Idebenone seems to improve the respiratory function in the long term. The trials with mRNA transcription, through nonsense mutations or exon 51 skipping, show some beneficial results in a few functional tests, but they are limited to a small set of DMD patients.


Assuntos
Distrofia Muscular de Duchenne/tratamento farmacológico , Corticosteroides/uso terapêutico , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Inibidores da Colinesterase/uso terapêutico , Distrofina/efeitos dos fármacos , Distrofina/metabolismo , Humanos , Distrofia Muscular de Duchenne/metabolismo
6.
Arq. neuropsiquiatr ; 77(8): 579-589, Aug. 2019. tab, graf
Artigo em Inglês | LILACS | ID: biblio-1019465

RESUMO

ABSTRACT In this review, we discuss the therapies used in the treatment of patients with Duchenne muscular dystrophy since the first description of the disease. A short description is given of the various theories based on disease pathogenesis, which give the substrates for the many therapeutic interventions. A brief review of the methods of evaluation used in therapeutic trials is made. Of all the treatments, the only drugs that are still considered able to modify the course of the disease are the corticosteroids (prednisone/prednisolone/deflazacort). Other drugs (coenzyme Q10 and creatine) have had a little effect in a few functions without adverse reactions. Idebenone seems to improve the respiratory function in the long term. The trials with mRNA transcription, through nonsense mutations or exon 51 skipping, show some beneficial results in a few functional tests, but they are limited to a small set of DMD patients.


RESUMO Nesta revisão são discutidas as terapêuticas empregadas no tratamento da distrofia muscular de Duchenne desde a descrição da doença. Apresentamos as diversas teorias que fundamentaram as intervenções terapêuticas, com uma breve descrição dos tipos de avaliação empregados nos ensaios terapêuticos. Dentre todos os tratamentos, a única medicação que até agora modificou o curso da doença foram os corticosteroides (prednisona/prednisolona/deflazacort). A coenzima Q10 e creatina tiveram algum efeito pequeno em algumas funções e evolução da doença sem efeitos colaterais. O idebenone mostrou efeito benéfico na função respiratória em longo prazo. As tentativas de intervir no gene da distrofina utilizando técnicas de transcrição do mRNA através das mutações sem sentido e técnicas que pulam o exon 51 mostram resultado muito discreto em algumas provas funcionais e limitado a uma parcela pequena de casos.


Assuntos
Humanos , Distrofia Muscular de Duchenne/tratamento farmacológico , Bloqueadores dos Canais de Cálcio/uso terapêutico , Inibidores da Colinesterase/uso terapêutico , Distrofina/efeitos dos fármacos , Distrofina/metabolismo , Corticosteroides/uso terapêutico , Distrofia Muscular de Duchenne/metabolismo , Anti-Inflamatórios/uso terapêutico , Antioxidantes/uso terapêutico
8.
Am J Pathol ; 187(11): 2520-2535, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28823869

RESUMO

The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.


Assuntos
Glucocorticoides/farmacologia , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular do Cíngulo dos Membros/tratamento farmacológico , Animais , Distrofina/efeitos dos fármacos , Distrofina/metabolismo , Glucocorticoides/administração & dosagem , Proteínas de Membrana/metabolismo , Camundongos , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/patologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Prednisona/administração & dosagem , Prednisona/farmacologia , Sarcoglicanas/efeitos dos fármacos , Sarcoglicanas/metabolismo
9.
Nat Commun ; 7: 10981, 2016 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-26964641

RESUMO

Carbohydrate-based infusion solutions are widely used in the clinic. Here we show that co-administration of phosphorodiamidate morpholino oligomers (PMOs) with glucose enhances exon-skipping activity in Duchenne muscular dystrophy (DMD) mdx mice. We identify a glucose-fructose (GF) formulation that potentiates PMO activity, completely corrects aberrant Dmd transcripts, restores dystrophin levels in skeletal muscles and achieves functional rescue without detectable toxicity. This activity is attributed to enhancement of GF-mediated PMO uptake in the muscle. We demonstrate that PMO cellular uptake is energy dependent, and that ATP from GF metabolism contributes to enhanced cellular uptake of PMO in the muscle. Collectively, we show that GF potentiates PMO activity by replenishing cellular energy stores under energy-deficient conditions in mdx mice. Our findings provide mechanistic insight into hexose-mediated oligonucleotide delivery and have important implications for the development of DMD exon-skipping therapy.


Assuntos
Distrofina/efeitos dos fármacos , Frutose/farmacologia , Glucose/farmacologia , Morfolinos/farmacologia , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , Animais , Distrofina/genética , Distrofina/metabolismo , Éxons , Terapia Genética , Hexoses/farmacologia , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , RNA Mensageiro/metabolismo
10.
Int J Nanomedicine ; 10: 5635-46, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26366082

RESUMO

In this study, we investigated a series of cationic polyelectrolytes (PEs) with different size and composition for their potential to improve delivery of an antisense phosphorodiamidate morpholino oligomer (PMO) both in vitro and in vivo. The results showed that the poly(diallyldimethylammonium chloride) (PDDAC) polymer series, especially PE-3 and PE-4, improves the delivery efficiency of PMO, comparable with Endoporter-mediated PMO delivery in vitro. The enhanced PMO delivery and targeting to dystrophin exon 23 was further observed in mdx mice, up to fourfold with the PE-4, compared with PMO alone. The cytotoxicity of the PEs was lower than that of Endoporter and polyethylenimine 25,000 Da in vitro, and was not clearly detected in muscle in vivo under the tested concentrations. Together, these results demonstrate that optimization of PE molecular size, composition, and distribution of cationic charge are key factors to achieve enhanced PMO exon-skipping efficiency. The increased efficiency and lower toxicity show this PDDAC series to be capable gene/antisense oligonucleotide delivery-enhancing agents for treating muscular dystrophy and other diseases.


Assuntos
Sistemas de Liberação de Medicamentos , Morfolinos/química , Oligonucleotídeos Antissenso/química , Poliaminas/química , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Distrofina/química , Distrofina/efeitos dos fármacos , Eletrodos , Éxons , Técnicas de Transferência de Genes , Camundongos , Camundongos Endogâmicos mdx , Microscopia Eletrônica de Transmissão , Morfolinos/farmacologia , Distrofias Musculares/tratamento farmacológico , Oligonucleotídeos Antissenso/farmacologia , Poliaminas/farmacologia , Polieletrólitos , Polietilenoimina/química , Polietilenoimina/farmacologia
12.
Nat Med ; 21(3): 270-5, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25642938

RESUMO

Antisense oligonucleotides (AONs) hold promise for therapeutic correction of many genetic diseases via exon skipping, and the first AON-based drugs have entered clinical trials for neuromuscular disorders. However, despite advances in AON chemistry and design, systemic use of AONs is limited because of poor tissue uptake, and recent clinical reports confirm that sufficient therapeutic efficacy has not yet been achieved. Here we present a new class of AONs made of tricyclo-DNA (tcDNA), which displays unique pharmacological properties and unprecedented uptake by many tissues after systemic administration. We demonstrate these properties in two mouse models of Duchenne muscular dystrophy (DMD), a neurogenetic disease typically caused by frame-shifting deletions or nonsense mutations in the gene encoding dystrophin and characterized by progressive muscle weakness, cardiomyopathy, respiratory failure and neurocognitive impairment. Although current naked AONs do not enter the heart or cross the blood-brain barrier to any substantial extent, we show that systemic delivery of tcDNA-AONs promotes a high degree of rescue of dystrophin expression in skeletal muscles, the heart and, to a lesser extent, the brain. Our results demonstrate for the first time a physiological improvement of cardio-respiratory functions and a correction of behavioral features in DMD model mice. This makes tcDNA-AON chemistry particularly attractive as a potential future therapy for patients with DMD and other neuromuscular disorders or with other diseases that are eligible for exon-skipping approaches requiring whole-body treatment.


Assuntos
Distrofina/efeitos dos fármacos , Coração/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular de Duchenne , Nanopartículas , Oligodesoxirribonucleotídeos Antissenso/farmacologia , RNA Mensageiro/análise , Animais , Barreira Hematoencefálica/metabolismo , Códon sem Sentido , Modelos Animais de Doenças , Distrofina/genética , Éxons , Terapia Genética , Camundongos , Microscopia Eletrônica de Transmissão , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Oligodesoxirribonucleotídeos Antissenso/metabolismo , Transcriptoma/efeitos dos fármacos
14.
Neuromuscul Disord ; 20(2): 102-10, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20079639

RESUMO

Duchenne muscular dystrophy (DMD) is caused by the lack of functional dystrophin protein, most commonly as a result of a range of out-of-frame mutations in the DMD gene. Modulation of pre-mRNA splicing with antisense oligonucleotides (AOs) to restore the reading frame has been demonstrated in vitro and in vivo, such that truncated but functional dystrophin is expressed. AO-induced skipping of exon 51 of the DMD gene, which could treat 13% of DMD patients, has now progressed to clinical trials. We describe here the methodical, cooperative comparison, in vitro (in DMD cells) and in vivo (in a transgenic mouse expressing human dystrophin), of 24 AOs of the phosphorodiamidate morpholino oligomer (PMO) chemistry designed to target exon 53 of the DMD gene, skipping of which could be potentially applicable to 8% of patients. A number of the PMOs tested should be considered worthy of development for clinical trial.


Assuntos
Distrofina/efeitos dos fármacos , Éxons/efeitos dos fármacos , Marcação de Genes/métodos , Terapia Genética/métodos , Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos Antissenso/farmacologia , Animais , Sequência de Bases/efeitos dos fármacos , Sequência de Bases/genética , Células Cultivadas , Modelos Animais de Doenças , Distrofina/química , Distrofina/genética , Éxons/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Morfolinas/química , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Morfolinos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Mutação/genética , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/uso terapêutico , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
15.
Toxicon ; 50(5): 589-99, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17572466

RESUMO

In a previous study, we showed that the Polybia paulista wasp venom causes strong myonecrosis. This study was undertaken to characterize the myotoxic potency of mastoparan (Polybia-MPII) isolated from venom (0.25 microg/microl) and injected in the tibial anterior (TA) muscle (i.m.) of Balb/c mice. The time course of the changes was followed at muscle degenerative (3 and 24h) and regenerative (3, 7, and 21 days) periods (n=6) after injection and compared to matched controls by calculation of the percentage of cross-sectional area affected and determination of creatine kinase (CK) activity (n=10). The results showed that although MP was strongly myotoxic, its capacity for regeneration was maintained high. Since the extent of tissue damage was not correlated with the CK serum levels, which remained very low, we raised the hypothesis that the enzyme underwent denaturation by the peptide. Evidence suggested that MP induced the death of TA fibers by necrosis and apoptosis and had the sarcolemma as its primordial target. Given its amphiphilic polycationic nature and based on the vast spectrum of functions attributed to the peptide, we suggest that MP interaction with cell membrane impaired the phosphorylation of dystrophin essential for sarcolemma mechanical stability, and disturbed Ca2+ mobilization with obvious implications on sarcoplasmic reticulum and mitochondrial functioning.


Assuntos
Músculo Esquelético/efeitos dos fármacos , Peptídeos/toxicidade , Venenos de Vespas/toxicidade , Vespas , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Cromatografia Líquida de Alta Pressão , Creatina Quinase/sangue , Distrofina/efeitos dos fármacos , Distrofina/metabolismo , Injeções Intramusculares , Peptídeos e Proteínas de Sinalização Intercelular , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Necrose , Peptídeos/síntese química , Fosforilação , Regeneração , Sarcolema/efeitos dos fármacos , Venenos de Vespas/síntese química
16.
Acta Myol ; 22(1): 15-21, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12966700

RESUMO

Aim of the study was to investigate whether the administration of gentamicin could restore dystrophin expression in striated muscles of patients with Duchenne muscular dystrophy caused by premature stop codon, as reported in mdx mice. Four Duchenne patients, still ambulant or in wheelchair stage for less than 4 months, selected among those with point mutations resulting in premature stop codons, received two 6-day cycles of gentamicin sulfate, at an interval of 7 weeks, according to the protocol approved by the Ethics Committee of the Second University of Naples. A muscle biopsy was performed after the second cycle of administration; the specimens were analysed by both immuno-histochemistry and Western blotting. Skeletal muscle changes were monitored by dynamic tests and Creatine Kinase values; at the beginning and end of treatment, cardiac and respiratory status was evaluated by electrocardiography, echocardiography, acoustic densitometry and vital capacity. Side-effects such as nephrotoxicity and ototoxicity were also monitored. Three out of four patients, who had the most permissive UGA as stop codon, showed positive results. In one patient, there was a dramatic re-expression of dystrophin by both immuno-histochemistry and Western blot; in two patients, dystrophin positive fibres were seen by the antibody to the rod domain with immuno-histochemistry; the fourth patient, with UAA as stop codon, showed no expression of dystrophin at all. These results suggest that gentamicin is able to recover dystrophin expression in a subset of Duchenne patients with nonsense mutations, raising the possibility of the first pharmacological treatment for muscular dystrophy.


Assuntos
Antibacterianos/uso terapêutico , Códon sem Sentido/efeitos dos fármacos , Códon sem Sentido/genética , Gentamicinas/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Antibacterianos/administração & dosagem , Criança , Pré-Escolar , Esquema de Medicação , Distrofina/análise , Distrofina/efeitos dos fármacos , Seguimentos , Gentamicinas/administração & dosagem , Humanos , Masculino , Músculo Esquelético/química , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/patologia , Fatores de Tempo , Resultado do Tratamento
17.
Acta Neuropathol ; 92(4): 331-40, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8891064

RESUMO

In this study we describe a novel experimental approach to quantify the relative susceptibility of (membrane-associated, contractile and mitochondrial) proteins in normal human muscle tissue sections to oxidative damage by the reactive oxygen species (ROS), hydroxyl (OH.) or superoxide (O2.-) radicals. The latter species were generated under controlled experimental conditions in vitro using a 60Co gamma radiation source, with subsequent analysis of damage to target proteins (dystrophin, beta-dystroglycan, beta-spectrin, fast and slow myosin heavy chain, NADH tetrazolium reductase, succinate dehydrogenase and cytochrome oxidase) via standard histochemistry, immunocytochemistry and electron microscopy of muscle tissue sections. In general terms, each of the proteins listed above was more susceptible to oxidative damage by OH., compared to O2.-. Different proteins (differing in structure, function or intracellular localisation) showed different susceptibility to oxidative damage, with certain mitochondrial proteins (succinate dehydrogenase, cytochrome oxidase) showing particular susceptibility. In addition, the use of monoclonal antibodies to four different regions of dystrophin showed the latter to contain both resistant and susceptible regions to ROS induced oxidative damage. At the ultrastructural level of subcellular organelle damage, mitochondria were identified as being particularly susceptible to ROS induced oxidative damage. We therefore speculate that oxidative damage to mitochondria and/or mitochondrial proteins may represent the principal initial route of free radical-induced damage within skeletal muscle tissue.


Assuntos
Proteínas Musculares/efeitos dos fármacos , Proteínas Musculares/imunologia , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Estresse Oxidativo/efeitos dos fármacos , Distrofina/efeitos dos fármacos , Radicais Livres/efeitos adversos , Humanos , Imuno-Histoquímica , Mitocôndrias/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/imunologia , Miosinas/efeitos dos fármacos , Espécies Reativas de Oxigênio/fisiologia , Espectrina/efeitos dos fármacos
18.
J Neurosci ; 16(12): 3791-7, 1996 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8656273

RESUMO

During synaptogenesis, agrin, released by motor nerves, causes the clustering of acetylcholine receptors (AChRs) in the skeletal muscle membrane. Although muscle alpha-dystroglycan has been postulated to be the receptor for the activity of agrin, previous experiments have revealed a discrepancy between the biological activity of soluble fragments of two isoforms of agrin produced by nerves and muscles, respectively, and their ability to bind alpha-dystroglycan. We have determined the specificity of the signaling receptor by investigating whether muscle agrin can block the activity of neural agrin on intact C2 myotubes. We find that a large excess of muscle agrin failed to inhibit either the number of AChR clusters or the phosphorylation of the AChR induced by picomolar concentrations of neural agrin. These results indicate that neural, but not muscle, agrin interacts with the signaling receptor. Muscle agrin did block the binding of neural agrin to isolated alpha-dystroglycan, however, suggesting either that alpha-dystroglycan is not the signaling receptor or that its properties in the membrane are altered. Direct assay of the binding of muscle or neural agrin to intact myotubes revealed only low-affinity binding. We conclude that the signaling receptor for agrin is a high-affinity receptor that is highly specific for the neural form.


Assuntos
Agrina/farmacologia , Fibras Musculares Esqueléticas/química , Agrina/química , Agrina/metabolismo , Animais , Ligação Competitiva/fisiologia , Linhagem Celular/química , Linhagem Celular/fisiologia , Colódio , Proteínas do Citoesqueleto/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Distroglicanas , Distrofina/efeitos dos fármacos , Distrofina/metabolismo , Isomerismo , Glicoproteínas de Membrana/efeitos dos fármacos , Glicoproteínas de Membrana/metabolismo , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/citologia , Junção Neuromuscular/química , Junção Neuromuscular/fisiologia , Fosforilação , Ratos , Receptores Colinérgicos/efeitos dos fármacos , Receptores Colinérgicos/metabolismo , Receptores Colinérgicos/ultraestrutura , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Sensibilidade e Especificidade , Transdução de Sinais/fisiologia , Torpedo
19.
Neuromuscul Disord ; 5(2): 105-14, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7767089

RESUMO

Human muscle cultures undergo a long-term loss of myotubes and a decline in dystrophin content, which can be prevented by glucocorticoid treatment of the cultures. We confirmed these findings and extended them to show that the utrophin content of control and dexamethasone-treated normal myotube cultures is not significantly different. In contrast to normal cultures, the utrophin content of long-term dexamethasone-treated DMD myotube cultures was significantly greater than that of the corresponding untreated cultures. Utrophin mRNA transcript levels normalized to total poly (A) were unaffected by dexamethasone treatment of either normal or DMD myotube cultures, suggesting the effect of dexamethasone on utrophin accumulation by DMD cultures is mediated post-transcriptionally. A combination of an increase in myotube numbers and lack of competition with dystrophin for membrane-binding sites in DMD myotubes may explain the distinct effects of dexamethasone on utrophin levels in normal and DMD cultures.


Assuntos
Proteínas do Citoesqueleto/efeitos dos fármacos , Glucocorticoides/farmacologia , Proteínas de Membrana , Fibras Musculares Esqueléticas/efeitos dos fármacos , Músculo Esquelético/citologia , Distrofias Musculares/metabolismo , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Dexametasona/farmacologia , Relação Dose-Resposta a Droga , Distrofina/efeitos dos fármacos , Humanos , Fibras Musculares Esqueléticas/metabolismo , RNA Mensageiro/metabolismo , Utrofina
20.
Neurology ; 43(2): 342-5, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8437700

RESUMO

Glucocorticoid therapy slows the progression of Duchenne muscular dystrophy. In muscle cultures, the addition of the glucocorticoid methylprednisolone increases myogenesis in most normal mixed and clonal cultures. Conversely, in some normal clonal and most dystrophic cultures, methylprednisolone inhibits fusion. However, in fusion-arrested normal and Becker muscular dystrophy cultures, dystrophin is expressed independently of fusion and of myosin heavy chain expression, and in some cases, expression is apparently enhanced by methylprednisolone. We suggest that dystrophin is a muscle-specific protein that does not require fusion for expression, and the methylprednisolone-induced enhancement of dystrophin expression may account for some of the clinical benefits of glucocorticoids in vivo.


Assuntos
Distrofina/efeitos dos fármacos , Metilprednisolona/farmacologia , Músculos/efeitos dos fármacos , Distrofias Musculares/metabolismo , Western Blotting , Células Cultivadas , Distrofina/análise , Humanos , Músculos/química , Músculos/patologia , Distrofias Musculares/patologia , Miosinas/análise , Miosinas/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...