Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Skelet Muscle ; 10(1): 30, 2020 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-33092650

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder stemming from a loss of functional dystrophin. Current therapeutic options for DMD are limited, as small molecule modalities remain largely unable to decrease the incidence or mitigate the consequences of repetitive mechanical insults to the muscle during eccentric contractions (ECCs). METHODS: Using a metabolomics-based approach, we observed distinct and transient molecular phenotypes in muscles of dystrophin-deficient MDX mice subjected to ECCs. Among the most chronically depleted metabolites was nicotinamide adenine dinucleotide (NAD), an essential metabolic cofactor suggested to protect muscle from structural and metabolic degeneration over time. We tested whether the MDX muscle NAD pool can be expanded for therapeutic benefit using two complementary small molecule strategies: provision of a biosynthetic precursor, nicotinamide riboside, or specific inhibition of the NAD-degrading ADP-ribosyl cyclase, CD38. RESULTS: Administering a novel, potent, and orally available CD38 antagonist to MDX mice successfully reverted a majority of the muscle metabolome toward the wildtype state, with a pronounced impact on intermediates of the pentose phosphate pathway, while supplementing nicotinamide riboside did not significantly affect the molecular phenotype of the muscle. However, neither strategy sustainably increased the bulk tissue NAD pool, lessened muscle damage markers, nor improved maximal hindlimb strength following repeated rounds of eccentric challenge and recovery. CONCLUSIONS: In the absence of dystrophin, eccentric injury contributes to chronic intramuscular NAD depletion with broad pleiotropic effects on the molecular phenotype of the tissue. These molecular consequences can be more effectively overcome by inhibiting the enzymatic activity of CD38 than by supplementing nicotinamide riboside. However, we found no evidence that either small molecule strategy is sufficient to restore muscle contractile function or confer protection from eccentric injury, undermining the modulation of NAD metabolism as a therapeutic approach for DMD.


Assuntos
Inibidores Enzimáticos/farmacologia , Metaboloma , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , NAD/metabolismo , Niacinamida/análogos & derivados , Compostos de Piridínio/farmacologia , ADP-Ribosil Ciclase 1/antagonistas & inibidores , Animais , Distrofina/deficiência , Inibidores Enzimáticos/uso terapêutico , Masculino , Glicoproteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Contração Muscular , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/fisiologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Compostos de Piridínio/uso terapêutico
2.
Biol Open ; 9(8)2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32718931

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease caused by mutation of the dystrophin gene. Pharmacological therapies that function independently of dystrophin and complement strategies aimed at dystrophin restoration could significantly improve patient outcomes. Previous observations have suggested that serotonin pathway modulation ameliorates dystrophic pathology, and re-application of serotonin modulators already used clinically would potentially hasten availability to DMD patients. In our study, we used dystrophin-deficient sapje and sapje-like zebrafish models of DMD for rapid and easy screening of several classes of serotonin pathway modulators as potential therapeutics. None of the candidate drugs tested significantly decreased the percentage of zebrafish exhibiting the dystrophic muscle phenotype in the short-term birefringence assay or lengthened the lifespan in the long-term survival assay. Although we did not identify an effective drug, we believe our data is of value to the DMD research community for future studies, and there is evidence that suggests serotonin modulation may still be a viable treatment strategy with further investigation. Given the widespread clinical use of selective serotonin reuptake inhibitors, tricyclic antidepressants and reversible inhibitors of monoamine oxidase, their reapplication to DMD is an attractive strategy in the field's pursuit to identify pharmacological therapies to complement dystrophin restoration strategies.


Assuntos
Distrofina/deficiência , Serotonina/metabolismo , Peixe-Zebra/metabolismo , Animais , Birrefringência , Avaliação Pré-Clínica de Medicamentos , Distrofina/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Receptores de Serotonina , Agonistas do Receptor de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Análise de Sobrevida
3.
Int J Mol Sci ; 20(23)2019 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-31801292

RESUMO

: Duchenne muscular dystrophy (DMD) is one of the most severe forms of inherited muscular dystrophies. The disease is caused by the lack of dystrophin, a structurally essential protein; hence, a definitive cure would necessarily have to pass through some form of gene and/or cell therapy. Cell- and genetic-based therapeutics for DMD have been explored since the 1990s and recently, two of the latter have been approved for clinical use, but their efficacy is still very low. In parallel, there have been great ongoing efforts aimed at targeting the downstream pathogenic effects of dystrophin deficiency using classical pharmacological approaches, with synthetic or biological molecules. However, as it is always the case with rare diseases, R&D costs for new drugs can represent a major hurdle for researchers and patients alike. This problem can be greatly alleviated by experimenting the use of molecules that had originally been developed for different conditions, a process known as drug repurposing or drug repositioning. In this review, we will describe the state of the art of such an approach for DMD, both in the context of clinical trials and pre-clinical studies.


Assuntos
Reposicionamento de Medicamentos/métodos , Distrofia Muscular de Duchenne/tratamento farmacológico , Fármacos Neuromusculares/uso terapêutico , Prednisona/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Distrofina/deficiência , Distrofina/genética , Gentamicinas/uso terapêutico , Humanos , Metformina/uso terapêutico , Camundongos Transgênicos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Pregnenodionas/uso terapêutico , Sinvastatina/uso terapêutico , Tadalafila/uso terapêutico , Tamoxifeno/uso terapêutico
4.
Acta Histochem ; 121(3): 268-276, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30642627

RESUMO

Dp71 is the major form of dystrophins (Dp) in the supraoptic nucleus (SON) and in the neural lobe of hypophysis (NL/HP). Dp71-null mice exhibit a hypo-osmolar status attributed to an altered osmosensitivity of the SON and to a perturbed vasopressinergic axis. Because oxytocin (OT) is implicated in osmoregulation via natriuresis, this study explored the oxytocinergic axis in Dp71-null mice after salt-loading (SL). Under normosmolar conditions, OT-mRNA expression was higher in the Dp71-null SON compared to wild-type (wt) and the OT peptide level has not changed. Dp-immunostaining was localized in astrocytes end-feet surrounding vessels in wt SON. This distribution changed in Dp71-null SON, Dp being detected in OT-soma of MCNs. nNOS and NADPH-diaphorase levels increased in the OT area of the Dp71-null SON compared to wt. In the NL/HP, OT level reduced in Dp71-null mice and Dp localization changed from pituicytes end-feet in wt SON to OT terminals in Dp71-null SON. Salt-Loading resulted in an increase of OT-mRNA and peptide levels in wt SON but had no effect in Dp71-null SON. In the NL/HP, OT content was reduced after SL. For Dp71-null mice, OT level, already low in control, was not modified by SL. Dp level was not affected by SL in the SON nor in the NL/HP. Our data confirmed the importance of Dp71 for the SON functionality in osmoregulation. The localization of Dp71 at the glial-vascular interface could be associated with SON osmosensitivity, leading to an adequate OT synthesis in the SON and release from the NL/HP upon plasmatic hyperosmolality.


Assuntos
Distrofina/deficiência , Hipotálamo/metabolismo , Osmorregulação/fisiologia , Ocitocina/metabolismo , Animais , Distrofina/metabolismo , Camundongos Knockout , NADPH Desidrogenase/metabolismo , Neurônios/metabolismo , Ocitocina/genética , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Supraóptico/metabolismo
5.
Cell Tissue Res ; 369(3): 591-602, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28623422

RESUMO

In Duchenne muscular dystrophy (DMD), lack of dystrophin leads to progressive muscle degeneration, with DMD patients suffering from cardiorespiratory failure. Cell therapy is an alternative to life-long corticoid therapy. Satellite cells, the stem cells of skeletal muscles, do not completely compensate for the muscle damage in dystrophic muscles. Elevated levels of proinflammatory and profibrotic factors, such as metalloproteinase 9 (MMP-9), impair muscle regeneration, leading to extensive fibrosis and poor results with myoblast transplantation therapies. Omega-3 is an anti-inflammatory drug that protects against muscle degeneration in the mdx mouse model of DMD. In the present study, we test our hypothesis that omega-3 affects MMP-9 and thereby benefits muscle regeneration and myoblast transplantation in the mdx mouse. We observe that omega-3 reduces MMP-9 gene expression and improves myoblast engraftment, satellite cell activation, and muscle regeneration by mechanisms involving, at least in part, the regulation of macrophages, as shown here with the fluorescence-activated cell sorting technique. The present study demonstrates the benefits of omega-3 on satellite cell survival and muscle regeneration, further supporting its use in clinical trials and cell therapies in DMD.


Assuntos
Distrofina/deficiência , Ácidos Graxos Ômega-3/farmacologia , Metaloproteinase 9 da Matriz/metabolismo , Fibras Musculares Esqueléticas/patologia , Mioblastos/enzimologia , Mioblastos/transplante , Células Satélites de Músculo Esquelético/patologia , Animais , Biomarcadores/metabolismo , Distrofina/metabolismo , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/efeitos dos fármacos , Atrofia Muscular/patologia , Mioblastos/efeitos dos fármacos , Necrose , Receptores Notch/metabolismo , Regeneração/efeitos dos fármacos , Células Satélites de Músculo Esquelético/efeitos dos fármacos , Células Satélites de Músculo Esquelético/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos
6.
Neurotherapeutics ; 14(2): 429-446, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27921261

RESUMO

Duchenne muscular dystrophy arises from the loss of dystrophin and is characterized by calcium dysregulation, muscular atrophy, and metabolic dysfunction. The secondary reduction of neuronal nitric oxide synthase (nNOS) from the sarcolemma reduces NO production and bioavailability. As NO modulates glucose uptake, metabolism, and mitochondrial bioenergetics, we investigated whether an 8-week nitrate supplementation regimen could overcome metabolic dysfunction in the mdx mouse. Dystrophin-positive control (C57BL/10) and dystrophin-deficient mdx mice were supplemented with sodium nitrate (85 mg/l) in drinking water. Following the supplementation period, extensor digitorum longus and soleus were excised and radioactive glucose uptake was measured at rest (basal) and during contraction. Gastrocnemius was excised and mitochondrial respiration was measured using the Oroboros Oxygraph. Tibialis anterior was analyzed immunohistochemically for the presence of dystrophin, nNOS, nitrotyrosine, IgG and CD45+ cells, and histologically to assess areas of damage and regeneration. Glucose uptake in the basal and contracting states was normal in unsupplemented mdx muscles but was reduced following nitrate supplementation in mdx muscles only. The mitochondrial utilization of substrates was also impaired in mdx gastrocnemius during phosphorylating and maximal uncoupled respiration, and nitrate could not improve respiration in mdx muscle. Although nitrate supplementation reduced mitochondrial hydrogen peroxide emission, it induced mitochondrial uncoupling in red gastrocnemius, increased muscle fiber peroxynitrite (nitrotyrosine), and promoted skeletal muscle damage. Our novel data suggest that despite lower nNOS protein expression and likely lower NO production in mdx muscle, enhancing NO production with nitrate supplementation in these mice has detrimental effects on skeletal muscle. This may have important relevance for those with DMD.


Assuntos
Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Nitratos/administração & dosagem , Óxido Nítrico Sintase Tipo I/deficiência , Animais , Distrofina/deficiência , Transporte de Elétrons , Glucose/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Mitocôndrias/fisiologia , Espécies Reativas de Oxigênio
8.
Hum Mol Genet ; 23(7): 1869-78, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24234649

RESUMO

Duchenne muscular dystrophy (DMD) is caused by a lack of the dystrophin protein and has no effective treatment at present. Zebrafish provide a powerful in vivo tool for high-throughput therapeutic drug screening for the improvement of muscle phenotypes caused by dystrophin deficiency. Using the dystrophin-deficient zebrafish, sapje, we have screened a total of 2640 compounds with known modes of action from three drug libraries to identify modulators of the disease progression. Six compounds that target heme oxygenase signaling were found to rescue the abnormal muscle phenotype in sapje and sapje-like, while upregulating the inducible heme oxygenase 1 (Hmox1) at the protein level. Direct Hmox1 overexpression by injection of zebrafish Hmox1 mRNA into fertilized eggs was found to be sufficient for a dystrophin-independent restoration of normal muscle via an upregulation of cGMP levels. In addition, treatment of mdx(5cv) mice with the PDE5 inhibitor, sildenafil, which was one of the six drugs impacting the Hmox1 pathway in zebrafish, significantly increased the expression of Hmox1 protein, thus making Hmox1 a novel target for the improvement of dystrophic symptoms. These results demonstrate the translational relevance of our zebrafish model to mammalian models and support the use of zebrafish to screen for new drugs to treat human DMD. The discovery of a small molecule and a specific therapeutic pathway that might mitigate DMD disease progression could lead to significant clinical implications.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Distrofina/genética , Heme Oxigenase-1/biossíntese , Distrofia Muscular de Duchenne/tratamento farmacológico , Animais , GMP Cíclico/biossíntese , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Distrofina/deficiência , Heme Oxigenase-1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Inibidores da Fosfodiesterase 5/farmacologia , Piperazinas/farmacologia , Purinas/farmacologia , RNA Mensageiro/genética , Transdução de Sinais/genética , Citrato de Sildenafila , Sulfonas/farmacologia , Regulação para Cima , Peixe-Zebra/genética
9.
PLoS One ; 8(7): e66617, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23843959

RESUMO

BACKGROUND: In Duchenne muscular dystrophy (DMD), loss of the membrane stabilizing protein dystrophin results in myofiber damage. Microinjury to dystrophic myofibers also causes secondary imbalances in sarcolemmic ion permeability and resting membrane potential, which modifies excitation-contraction coupling and increases proinflammatory/apoptotic signaling cascades. Although glucocorticoids remain the standard of care for the treatment of DMD, there is a need to investigate the efficacy of other pharmacological agents targeting the involvement of imbalances in ion flux on dystrophic pathology. METHODOLOGY/PRINCIPAL FINDINGS: We designed a preclinical trial to investigate the effects of lansoprazole (LANZO) administration, a proton pump inhibitor, on the dystrophic muscle phenotype in dystrophin deficient (mdx) mice. Eight to ten week-old female mice were assigned to one of four treatment groups (n = 12 per group): (1) vehicle control; (2) 5 mg/kg/day LANZO; (3) 5 mg/kg/day prednisolone; and (4) combined treatment of 5 mg/kg/day prednisolone (PRED) and 5 mg/kg/day LANZO. Treatment was administered orally 5 d/wk for 3 months. At the end of the study, behavioral (Digiscan) and functional outcomes (grip strength and Rotarod) were assessed prior to sacrifice. After sacrifice, body, tissue and organ masses, muscle histology, in vitro muscle force, and creatine kinase levels were measured. Mice in the combined treatment groups displayed significant reductions in the number of degenerating muscle fibers and number of inflammatory foci per muscle field relative to vehicle control. Additionally, mice in the combined treatment group displayed less of a decline in normalized forelimb and hindlimb grip strength and declines in in vitro EDL force after repeated eccentric contractions. CONCLUSIONS/SIGNIFICANCE: Together our findings suggest that combined treatment of LANZO and prednisolone attenuates some components of dystrophic pathology in mdx mice. Our findings warrant future investigation of the clinical efficacy of LANZO and prednisolone combined treatment regimens in dystrophic pathology.


Assuntos
Distrofina/genética , Lansoprazol/farmacologia , Força Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Inibidores da Bomba de Prótons/farmacologia , Animais , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Distrofina/deficiência , Feminino , Expressão Gênica , Glucocorticoides/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Prednisolona/farmacologia
10.
Clin Nutr ; 32(3): 472-5, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22727548

RESUMO

BACKGROUND & AIMS: Duchenne muscular dystrophy (DMD) is a genetic muscle disease caused by the absence of dystrophin. An established animal model of DMD is the mdx mouse, which is unable to express dystrophin. Inflammation, particularly the proinflammatory cytokine tumor necrosis factor alpha (TNF-α), strongly contributes to necrosis in the dystrophin-deficient fibers of the mdx mice and in DMD. In this study we investigated whether the antioxidant N-acetylcysteine (NAC) decreases TNF-α levels and protects the diaphragm muscle of mdx mice against necrosis. METHODS: Mdx mice (14 days old) received daily intraperitoneal injections of NAC for 14 days, followed by removal of the diaphragm muscle. Control mdx mice were injected with saline. RESULTS: NAC reduced TNF-α and 4-HNE-protein adducts levels, inflammation, creatine kinase levels, and myonecrosis in diaphragm muscle. CONCLUSIONS: NAC may be used as a complementary treatment for dystrophinopathies. However, clinical trials are needed to determine the appropriate dose for patients with Duchenne muscular dystrophy.


Assuntos
Acetilcisteína/uso terapêutico , Diafragma/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Necrose/tratamento farmacológico , Fator de Necrose Tumoral alfa/sangue , Animais , Antioxidantes/uso terapêutico , Diafragma/patologia , Distrofina/deficiência , Distrofina/genética , Inflamação/tratamento farmacológico , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Músculo Esquelético/patologia , Necrose/patologia
11.
PLoS One ; 7(4): e34204, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22509280

RESUMO

BACKGROUND: Dmd(mdx) (mdx) mice are used as a genetic and biochemical model of dystrophin deficiency. The long-term consequences of glucocorticoid (GC) treatment on dystrophin-deficient skeletal and heart muscle are not yet known. Here we used systematic phenotyping to assess the long-term consequences of GC treatment in mdx mice. Our investigation addressed not only the effects of GC on the disease phenotype but also the question of whether GCs can be used as a positive control for preclinical drug evaluations. METHODS AND FINDINGS: We performed nine pre-clinical efficacy trials (treated N = 129, untreated N = 106) of different durations in 9-to-50-week-old dystrophic mdx mice over a 3-year time period using standardized methods. In all these trials, we used either 1 mg/kg body weight of prednisone or 5 mg/kg body weight of prednisolone as positive controls to compare the efficacy of various test drugs. Data from untreated controls and GC-treated mice in the various trials have been pooled and analyzed to assess the effects of GCs on dystrophin-deficient skeletal and cardiac muscles of mdx mice. Our results indicate that continuous GC treatment results in early (e.g., at 50 days) improvements in normalized parameters such as grip strength, motor coordination and maximal in vitro force contractions on isolated EDL muscle, but these initial benefits are followed by a progressive loss of muscle strength after 100 days. We also found a significant increase in heart fibrosis that is reflected in a significant deterioration in cardiac systolic function after 100 days of treatment. CONCLUSION: Continuous administration of prednisone to mdx mice initially improves skeletal muscle strength, but further therapy result in deterioration of muscle strength and cardiac function associated with enhanced cardiac fibrosis. These results suggest that GCs may not serve as an appropriate positive control for long-term mdx mouse preclinical trials.


Assuntos
Avaliação Pré-Clínica de Medicamentos/normas , Glucocorticoides/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Distrofina/deficiência , Feminino , Fibrose/induzido quimicamente , Glucocorticoides/administração & dosagem , Força da Mão , Coração/efeitos dos fármacos , Coração/fisiologia , Camundongos , Camundongos Endogâmicos mdx , Atividade Motora/efeitos dos fármacos , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiologia , Miocárdio/metabolismo , Miocárdio/patologia , Padrões de Referência , Fatores de Tempo
12.
PLoS One ; 5(5): e10763, 2010 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-20505827

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common, lethal disease of childhood. One of 3500 new-born males suffers from this universally-lethal disease. Other than the use of corticosteroids, little is available to affect the relentless progress of the disease, leading many families to use dietary supplements in hopes of reducing the progression or severity of muscle wasting. Arginine is commonly used as a dietary supplement and its use has been reported to have beneficial effects following short-term administration to mdx mice, a genetic model of DMD. However, the long-term effects of arginine supplementation are unknown. This lack of knowledge about the long-term effects of increased arginine metabolism is important because elevated arginine metabolism can increase tissue fibrosis, and increased fibrosis of skeletal muscles and the heart is an important and potentially life-threatening feature of DMD. METHODOLOGY: We use both genetic and nutritional manipulations to test whether changes in arginase metabolism promote fibrosis and increase pathology in mdx mice. Our findings show that fibrotic lesions in mdx muscle are enriched with arginase-2-expressing macrophages and that muscle macrophages stimulated with cytokines that activate the M2 phenotype show elevated arginase activity and expression. We generated a line of arginase-2-null mutant mdx mice and found that the mutation reduced fibrosis in muscles of 18-month-old mdx mice, and reduced kyphosis that is attributable to muscle fibrosis. We also observed that dietary supplementation with arginine for 17-months increased mdx muscle fibrosis. In contrast, arginine-2 mutation did not reduce cardiac fibrosis or affect cardiac function assessed by echocardiography, although 17-months of dietary supplementation with arginine increased cardiac fibrosis. Long-term arginine treatments did not decrease matrix metalloproteinase-2 or -9 or increase the expression of utrophin, which have been reported as beneficial effects of short-term treatments. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that arginine metabolism by arginase promotes fibrosis of muscle in muscular dystrophy and contributes to kyphosis. Our findings also show that long-term, dietary supplementation with arginine exacerbates fibrosis of dystrophic heart and muscles. Thus, commonly-practiced dietary supplementation with arginine by DMD patients has potential risk for increasing pathology when performed for long periods, despite reports of benefits acquired with short-term supplementation.


Assuntos
Arginina/metabolismo , Macrófagos/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Animais , Arginase/metabolismo , Arginina/administração & dosagem , Arginina/farmacologia , Cardiomiopatia Dilatada/enzimologia , Cardiomiopatia Dilatada/patologia , Citocinas/metabolismo , Distrofina/deficiência , Distrofina/metabolismo , Fibrose , Deleção de Genes , Inflamação/complicações , Inflamação/enzimologia , Inflamação/patologia , Cifose/complicações , Cifose/enzimologia , Cifose/patologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/complicações , Distrofia Muscular Animal/enzimologia , Óxido Nítrico Sintase Tipo I/metabolismo , Transporte Proteico/efeitos dos fármacos , Células Th2/efeitos dos fármacos
13.
Neuroscience ; 166(4): 1129-39, 2010 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-20109538

RESUMO

The dystrophin defective mdx mouse, acknowledged model of Duchenne Muscular Dystrophy (DMD), bears outstanding alterations of the cortical architecture, that could be responsible for the cognitive impairment often accompanying this pathological condition. Using a retrograde tract tracing technique to label neurons in Golgi-like fashion, we investigated the fine anatomical organization of associative cortico-cortical projections in mdx mice. While the absolute number of associative pyramidal neurons was significantly higher in mdx than in control animals, the ratio between the number of supra- and infragranular cortico-cortical cells was substantially unmodified. Basal dendrites of layer 2/3 pyramidal neurons displayed longer terminal branches in mdx compared to controls. Finally, the density of dendritic spines was significantly lower in mdx animals. The anomalies of associative cortico-cortical projections provide potential groundwork on the neurobiological bases of cognitive involvement in DMD and value the role of cortical microcircuitry alterations as possible source of interference with peripheral motor impairment.


Assuntos
Córtex Cerebral/patologia , Transtornos Cognitivos/patologia , Distrofina/deficiência , Distrofia Muscular de Duchenne/patologia , Rede Nervosa/patologia , Células Piramidais/patologia , Animais , Biotina/análogos & derivados , Contagem de Células , Forma Celular/fisiologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Transtornos Cognitivos/etiologia , Transtornos Cognitivos/metabolismo , Dendritos/metabolismo , Dendritos/patologia , Dendritos/ultraestrutura , Espinhas Dendríticas/metabolismo , Espinhas Dendríticas/patologia , Espinhas Dendríticas/ultraestrutura , Dextranos , Modelos Animais de Doenças , Distrofina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Córtex Motor/metabolismo , Córtex Motor/patologia , Córtex Motor/fisiopatologia , Distrofia Muscular de Duchenne/complicações , Distrofia Muscular de Duchenne/metabolismo , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Marcadores do Trato Nervoso , Células Piramidais/metabolismo , Células Piramidais/ultraestrutura , Córtex Somatossensorial/metabolismo , Córtex Somatossensorial/patologia , Córtex Somatossensorial/fisiopatologia , Coloração e Rotulagem
14.
J Neurosci Res ; 88(2): 324-34, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19658196

RESUMO

Patients with Duchenne muscular dystrophy (DMD) and mdx mice, devoid of dystrophin proteins, show altered ionic homeostasis. To clarify dystrophin's involvement in the central control of osmotic stimuli, we investigated the effect of the disruption of Dp71, the major form of dystrophin in the brain, on the hypothalamoneurohypophysis system (HNHS) osmoregulatory response. Dp71 and Dp140 are the principal DMD gene products in the supraoptic nucleus (SON) and neurohypophysis (NH). They are present in astrocyte and pituicyte end-feet, suggesting involvement in both intrinsic osmosensitivity of the SON and vasopressin (AVP) release from the NH. In Dp71-null mice, the cellular distribution of Dp140 was modified, this protein being detected on the membrane of magnocellular soma. The plasma osmolality of Dp71-null mice was lower than that of wild-type mice under normal conditions, and this difference was maintained after salt loading, indicating a change in the set point for osmoregulation in the absence of Dp71. The increase in AVP levels detected in the SON and NH of the wild-type was not observed in Dp71-null mice following salt loading, and the increase in AVP mRNA levels in the SON was smaller in Dp71-null than in wild-type mice. This suggests that Dp71 may be involved in the functional activity of the HNHS. Its astrocyte end-feet localization emphasizes the importance of neuronal-vascular-glial interactions for the central detection of osmolality. In the SON, Dp71 may be involved in osmosensitivity and definition of the "osmostat," whereas, in the neurohypophysis, it may be involved in fine-tuning AVP release.


Assuntos
Encéfalo/fisiologia , Distrofina/metabolismo , Hipotálamo/fisiologia , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Sangue/metabolismo , Distrofina/deficiência , Distrofina/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neuro-Hipófise/fisiologia , RNA Mensageiro/metabolismo , Sais/metabolismo , Núcleo Supraóptico/fisiologia , Vasopressinas/metabolismo , Água/metabolismo
15.
Rev. neurol. (Ed. impr.) ; 49(7): 369-375, 1 oct., 2009. ilus, tab
Artigo em Espanhol | IBECS | ID: ibc-94838

RESUMO

Introducción y desarrollo. La distrofia muscular de Duchenne (DMD) es una de las enfermedades genéticas neuromusculares de mayor gravedad y frecuencia en niños. Es una enfermedad discapacitante que ocasiona un deterioro progresivo de los músculos y lleva al paciente a la muerte en la mayoría de los casos por problemas cardiorrespiratorios. Hasta hace algún tiempo, la calidad y expectativa de vida de los pacientes eran reducidas y las opciones terapéuticas limitadas; sin embargo, recientemente se ha establecido un conjunto de intervenciones que modifica de manera significativa el progreso de la enfermedad y la calidad de vida de los pacientes con DMD. Se están desarrollando diversos enfoques terapéuticos para corregir molecularmente el defecto genético en estos pacientes. Mientras esto ocurre, es necesario implementar sistemas coordinados para mantener en la mejor condición física posible a los pacientes. Conclusiones. La detección temprana de las complicaciones permite identificar a los pacientes para canalizarlos a un tratamiento adecuado; sin embargo, esto depende del diagnóstico y seguimiento oportuno. La realización de estas intervenciones involucra a un grupo multidisciplinario de alta especialidad y necesita la colaboración del paciente y de su familia (AU)


Introduction and development. Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease of genetic origin that affects male children. It is characterized by progressive muscle deterioration which results in the patient becoming wheelchair-dependent until death from cardio-respiratory complications. A few years ago, DMD patients’ life quality and expectancy were poor and treatment options limited; valuable recommendations that significantly delay the progress of the disease and improve the patient’s life quality have been brought about recently. Numerous therapeutic approaches are now in development in order to correct the DMD genetic defect at molecular level. In the mean time, a comprehensive system to maintain patients in their best possible physical condition is needed. Conclusions. Accurate detection of complications enables caregivers to determine which patients are at higher risk and to provide treatment accordingly. Nevertheless, all of these efforts are dependent on early clinical and molecular diagnosis, careful record of clinical changes and long-term followup of DMD patients. Furthermore, the involvement of multidisciplinary groups and the patient’s family is essential in said interventions (AU)


Assuntos
Humanos , Distrofia Muscular de Duchenne/genética , Esteroides/uso terapêutico , Fenótipo , Distrofina/deficiência , Mutação/genética , Qualidade de Vida , Técnicas de Exercício e de Movimento , Posicionamento do Paciente , Exercícios Respiratórios
16.
Hum Mol Genet ; 18(21): 4089-101, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19648295

RESUMO

Duchenne Muscular Dystrophy is an inherited muscle degeneration disease for which there is still no efficient treatment. However, compounds active on the disease may already exist among approved drugs but are difficult to identify in the absence of cellular models. We used the Caenorhabditis elegans animal model to screen a collection of 1000 already approved compounds. Two of the most active hits obtained were methazolamide and dichlorphenamide, carbonic anhydrase inhibitors widely used in human therapy. In C. elegans, these drugs were shown to interact with CAH-4, a putative carbonic anhydrase. The therapeutic efficacy of these compounds was further validated in long-term experiments on mdx mice, the mouse model of Duchenne Muscular Dystrophy. Mice were treated for 120 days with food containing methazolamide or dichlorphenamide at two doses each. Musculus tibialis anterior and diaphragm muscles were histologically analyzed and isometric muscle force was measured in M. extensor digitorum longus. Both substances increased the tetanic muscle force in the treated M. extensor digitorum longus muscle group, dichlorphenamide increased the force significantly by 30%, but both drugs failed to increase resistance of muscle fibres to eccentric contractions. Histological analysis revealed a reduction of centrally nucleated fibers in M. tibialis anterior and diaphragm in the treated groups. These studies further demonstrated that a C. elegans-based screen coupled with a mouse model validation strategy can lead to the identification of potential pharmacological agents for rare diseases.


Assuntos
Inibidores da Anidrase Carbônica/farmacologia , Modelos Animais de Doenças , Distrofina/deficiência , Distrofia Muscular Animal/prevenção & controle , Animais , Caenorhabditis elegans/efeitos dos fármacos , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Inibidores da Anidrase Carbônica/metabolismo , Anidrases Carbônicas/genética , Anidrases Carbônicas/metabolismo , Diclorofenamida/farmacologia , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Distrofina/genética , Humanos , Metazolamida/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Atividade Motora , Contração Muscular/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Interferência de RNA , Fatores de Tempo
17.
Int Rev Cytol ; 240: 1-30, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15548414

RESUMO

Duchenne muscular dystrophy (DMD), a severe X-linked genetic disease affecting one in 3500 boys, is the most common myopathy in children. DMD is due to a lack of dystrophin, a submembrane protein of the cytoskeleton, which leads to the progressive degeneration of skeletal, cardiac, and smooth muscle tissue. A milder form of the disease, Becker muscular dystrophy (BMD), is characterized by the presence of a semifunctional truncated dystrophin, or reduced levels of full-length dystrophin. DMD is the focus of three different supportive or therapeutic approaches: gene therapy, cell therapy, and drug therapy. Here we consider these approaches in terms of three potential goals: improvement of dystrophic phenotype, expression of dystrophin, and overexpression of utrophin. Utrophin exhibits 80% homology with dystrophin and is able to perform similar functions. Pharmacological strategies designed to overexpress utrophin appear promising and may circumvent many obstacles to gene and cell-based therapies.


Assuntos
Distrofina/deficiência , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Duchenne/terapia , Animais , Avaliação Pré-Clínica de Medicamentos/tendências , Distrofina/genética , Terapia Genética/tendências , Humanos , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Utrofina/genética , Utrofina/metabolismo
18.
Neuromuscul Disord ; 14(6): 365-70, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15145337

RESUMO

Duchenne muscular dystrophy is a degenerative muscular disease caused by mutations in the dystrophin gene. There is no curative treatment against Duchenne muscular dystrophy. In several countries, the steroid prednisone (or analogs) is prescribed as a palliative treatment. In the model animal Caenorhabditis elegans, mutations of the dys-1 dystrophin-like gene lead to a muscular degenerative phenotype when they are associated with a mild MyoD mutation. This cheap and fast-growing model of dystrophinopathy may be used to screen for molecules able to slow muscle degeneration. In a blind screen of approximately 100 compounds covering a wide spectrum of targets, we found that prednisone is beneficial to the C. elegans dystrophin-deficient muscles. Prednisone reduces by 40% the number of degenerating cells in this animal. This result is a proof-of-principle for the use of C. elegans as a tool in the search for molecules active against the effects of dystrophin-deficiency. Moreover, since C. elegans is not susceptible to inflammation, this suggests that prednisone exerts a direct effect on muscle survival.


Assuntos
Distrofina/deficiência , Distrofia Muscular de Duchenne/tratamento farmacológico , Prednisona/uso terapêutico , Animais , Anti-Inflamatórios/uso terapêutico , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Distrofina/genética , Distrofina/metabolismo , Proteínas Musculares/deficiência , Proteínas Musculares/genética , Músculos/efeitos dos fármacos , Distrofia Muscular de Duchenne/metabolismo , Mutação , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Fatores de Tempo , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
19.
Am J Chin Med ; 29(2): 281-92, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11527070

RESUMO

The purpose of this study was to access the effects of chinese herbal medicines on Duchenne muscular dystrophy (DMD). We use dystrophin-deficient mice (mdx), an animal model of DMD, to evaluate the effect of chinese herbal medicines on locomotor activity. The consumption of water for each mouse was controlled during the three-month experimental session. Each mouse was allowed to drink 3 ml water with or without herbal medicines daily for three months. The estimated intake of chinese herbal medicine in adult mdx mouse with 30 g weight is 100 mg/kg per day, close to a dose used in human. The locomotor activity of the mdx mice was measured every month. Monitoring the locomotor activity of mdx mice after three-month administration of chinese herbal medicines, the results showed that liu-wei-di-huang-wan (LDW) and san-lin-pai-tsu-san (SPS) can facilitate locomotor activity with the parameters of horizontal activity, total distance, number of movements, movement time, vertical activity, number of vertical movements, vertical movement time, stereotypy, number of stereotyped movements, and stereotyped movement time. These results suggest that either LDW or SPS can act as a potent herbal medicine for the pharmacological treatment of DMD patients.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Movimento/efeitos dos fármacos , Distrofia Muscular de Duchenne/tratamento farmacológico , Análise de Variância , Animais , Progressão da Doença , Distrofina/deficiência , Masculino , Camundongos , Camundongos Endogâmicos mdx , Modelos Animais , Atividade Motora/efeitos dos fármacos , Debilidade Muscular/prevenção & controle , Distrofia Muscular de Duchenne/fisiopatologia , Qualidade de Vida , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA