Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
1.
FASEB J ; 35(12): e22034, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34780665

RESUMO

Mutation to the gene encoding dystrophin can cause Duchenne muscular dystrophy (DMD) and increase the sensitivity to stress in vertebrate species, including the mdx mouse model of DMD. Behavioral stressors can exacerbate some dystrophinopathy phenotypes of mdx skeletal muscle and cause hypotension-induced death. However, we have discovered that a subpopulation of mdx mice present with a wildtype-like response to mild (forced downhill treadmill exercise) and moderate (scruff restraint) behavioral stressors. These "stress-resistant" mdx mice are more physically active, capable of super-activating the hypothalamic-pituitary-adrenal and renin-angiotensin-aldosterone pathways following behavioral stress and they express greater levels of mineralocorticoid and glucocorticoid receptors in striated muscle relative to "stress-sensitive" mdx mice. Stress-resistant mdx mice also presented with a less severe striated muscle histopathology and greater exercise and skeletal muscle oxidative capacity at rest. Most interestingly, female mdx mice were more physically active following behavioral stressors compared to male mdx mice; a response abolished after ovariectomy and rescued with estradiol. We demonstrate that the response to behavioral stress greatly impacts disease severity in mdx mice suggesting the management of stress in patients with DMD be considered as a therapeutic approach to ameliorate disease progression.


Assuntos
Comportamento Animal , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/patologia , Condicionamento Físico Animal , Estresse Psicológico/complicações , Animais , Modelos Animais de Doenças , Distrofina/deficiência , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Knockout , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/psicologia , Distrofia Muscular de Duchenne/etiologia , Distrofia Muscular de Duchenne/psicologia , Fatores Sexuais
2.
Exp Cell Res ; 406(2): 112766, 2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34364881

RESUMO

Duchene muscular dystrophy leads to progressive muscle structural and functional decline due to chronic degenerative-regenerative cycles. Enhancing the regenerative capacity of dystrophic muscle provides potential therapeutic options. We previously demonstrated that the circadian clock repressor Rev-erbα inhibited myogenesis and Rev-erbα ablation enhanced muscle regeneration. Here we show that Rev-erbα deficiency in the dystrophin-deficient mdx mice promotes regenerative myogenic response to ameliorate muscle damage. Loss of Rev-erbα in mdx mice improved dystrophic pathology and muscle wasting. Rev-erbα-deficient dystrophic muscle exhibit augmented myogenic response, enhanced neo-myofiber formation and attenuated inflammatory response. In mdx myoblasts devoid of Rev-erbα, myogenic differentiation was augmented together with up-regulation of Wnt signaling and proliferative pathways, suggesting that loss of Rev-erbα inhibition of these processes contributed to the improvement in regenerative myogenesis. Collectively, our findings revealed that the loss of Rev-erbα function protects dystrophic muscle from injury by promoting myogenic repair, and inhibition of its activity may have therapeutic utilities for muscular dystrophy.


Assuntos
Diferenciação Celular , Músculo Esquelético/citologia , Distrofia Muscular Animal/prevenção & controle , Distrofia Muscular de Duchenne/prevenção & controle , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/antagonistas & inibidores , Regeneração , Animais , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/etiologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Via de Sinalização Wnt
3.
FASEB J ; 35(6): e21628, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33982338

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal genetic muscle disorder caused by recessive mutations in dystrophin gene, affecting 1/3000 males. Gene therapy has been proven to ameliorate dystrophic pathology. To investigate therapeutic benefits from long-term effect of human mini-dystrophin and functional outcomes, transgenic mdx mice (Tg-mdx) containing a single copy of human mini-dystrophin (∆hDys3849) gene, five rods (Rods1-2, Rods22-24), and two hinges (H1 and H4) driven by a truncated creatine-kinase promoter (dMCK) in a recombinant adeno-associated viral vector (rAAV) backbone, were generated and used to determine gene expression and improvement of muscle function. Human mini-dystrophin gene expression was found in a majority of the skeletal muscles, but no expression in cardiac muscle. Dystrophin-associated glycoproteins (DAGs) such as sarcoglycans and nNOS were restored at the sarcolemma and coincided with human mini-dystrophin gene expression at the ages of 6, 10, and 20 months; Morphology of dystrophic muscle expressing the human mini-dystrophin gene was improved and central nuclei were reduced. Myofiber membrane integrity was improved by Evans blue dye test. Improvement in treadmill running and grip force was observed in transgenic mice at 6 months. Tetanic force and specific force of tibialis anterior (TA) muscle were significantly increased at the ages of 6, 10, and 20 months. Pseudohypertrophy was not found in TA muscle at 10 and 20 months when compared with wild-type C57 (WT) group. This study demonstrated that the long-term effects of human mini-dystrophin effectively ameliorated pathology and improved the functions of the dystrophic muscles in the transgenic DMD mouse model.


Assuntos
Distrofina/metabolismo , Terapia Genética , Músculo Esquelético/fisiologia , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Animais , Distrofina/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/citologia , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/etiologia , Distrofia Muscular de Duchenne/patologia
4.
Pol J Vet Sci ; 20(2): 387-394, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28865216

RESUMO

The aim of this study was to explain the correlations between selenium deficiency, hemostatic and biochemical disorders, and the progression of pathological changes in calves diagnosed with nutritional muscular dystrophy (NMD). The study was performed on 20 calves with supplementation of 8 ml selenium and vitamin E preparation and 20 calves with symptoms of NMD. Blood was sampled from calves aged 5, 12 and 19 days. On day 19, samples of the biceps femoris muscle were collected from 6 animals in each group for histopathological analysis. The following blood parameters were determined: PLT, PT, TT, APTT, fibrinogen and D-dimer concentrations, antithrombin III activity, glucose, selenium and vitamin E concentrations, activity of CK, LDH and GSH-Px. Muscle sections were stained with H&E and HBFP. Platelet counts were significantly lower in calves with symptoms of NMD. No significant differences in coagulation parameters were observed between the groups. Sick calves were diagnosed with hyperglycemia and elevation of CK and LDH activity. Selenium and vitamin E concentrations in the blood serum were significantly lower in the experimental group together with significant drop in GSH-Px activity. Changes characteristic of Zenker's necrosis were observed in a muscle of the sick animals. To our best knowledge this is the first study in which the attempt was made to explain the relationship between selenium deficiency and changes in the coagulation system in ruminants.


Assuntos
Transtornos da Coagulação Sanguínea/veterinária , Doenças dos Bovinos/sangue , Músculo Esquelético/patologia , Distrofia Muscular Animal/etiologia , Distúrbios Nutricionais/veterinária , Selênio/deficiência , Animais , Transtornos da Coagulação Sanguínea/etiologia , Transtornos da Coagulação Sanguínea/patologia , Bovinos , Doenças dos Bovinos/patologia , Glutationa Peroxidase/metabolismo , Distrofia Muscular Animal/sangue , Distrofia Muscular Animal/patologia , Distúrbios Nutricionais/sangue , Distúrbios Nutricionais/etiologia , Distúrbios Nutricionais/patologia , Vitamina E/metabolismo
5.
J Pathol ; 239(1): 10-22, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26800321

RESUMO

Injury to skeletal muscle, whether acute or chronic, triggers macrophage-mediated innate immunity in a manner which can be either beneficial or harmful for subsequent repair. Endogenous ligands for Toll-like receptor 2 (TLR2) are released by damaged tissues and might play an important role in activating the innate immune system following muscle injury. To test this hypothesis, we compared macrophage behaviour and muscle repair mechanisms in mice lacking TLR2 under conditions of either acute (cardiotoxin-induced) or chronic (mdx mouse genetic model of Duchenne muscular dystrophy; DMD) muscle damage. In previously healthy muscle subjected to acute damage, TLR2 deficiency reduced macrophage numbers in the muscle post-injury but did not alter the expression pattern of the prototypical macrophage polarization markers iNOS and CD206. In addition, there was abnormal persistence of necrotic fibres and impaired regeneration in TLR2-/- muscles after acute injury. In contrast, TLR2 ablation in chronically diseased muscles of mdx mice not only resulted in significantly reduced macrophage numbers but additionally modified their phenotype by shifting from inflammatory (iNOS(pos) CD206(neg) ) to more anti-inflammatory (iNOS(neg) CD206(pos) ) characteristics. This decrease in macrophage-mediated inflammation was associated with ameliorated muscle histopathology and improved force-generating capacity of the dystrophic muscle. Our results suggest that the role of TLR2 in macrophage function and skeletal muscle repair depends greatly upon the muscle injury context, and raise the possibility that inhibition of TLR2 could serve as a useful therapeutic measure in DMD.


Assuntos
Músculo Esquelético/lesões , Distrofia Muscular Animal/etiologia , Distrofia Muscular de Duchenne/etiologia , Receptor 2 Toll-Like/deficiência , Cicatrização/fisiologia , Análise de Variância , Animais , Cardiotoxinas/toxicidade , Células Cultivadas , Diafragma/fisiologia , Modelos Animais de Doenças , Feminino , Lectinas Tipo C/metabolismo , Ativação de Macrófagos/fisiologia , Masculino , Receptor de Manose , Lectinas de Ligação a Manose/metabolismo , Camundongos Endogâmicos mdx , Fibras Musculares Esqueléticas/fisiologia , Força Muscular/fisiologia , Músculo Esquelético/fisiologia , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores de Superfície Celular/metabolismo
6.
Neuromuscul Disord ; 25(5): 363-70, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25813339

RESUMO

Animal models of dystrophin deficient muscular dystrophy, most notably canine X-linked muscular dystrophy, play an important role in developing new therapies for human Duchenne muscular dystrophy. Although the canine disease is a model of the human disease, the variable severity of clinical presentations in the canine may be problematic for pre-clinical trials, but also informative. Here we describe a family of Labrador Retrievers with three generations of male dogs having markedly increased serum creatine kinase activity, absence of membrane dystrophin, but with undetectable clinical signs of muscle weakness. Clinically normal young male Labrador Retriever puppies were evaluated prior to surgical neuter by screening laboratory blood work, including serum creatine kinase activity. Serum creatine kinase activities were markedly increased in the absence of clinical signs of muscle weakness. Evaluation of muscle biopsies confirmed a dystrophic phenotype with both degeneration and regeneration. Further evaluations by immunofluorescence and western blot analysis confirmed the absence of muscle dystrophin. Although dystrophin was not identified in the muscles, we did not find any detectable deletions or duplications in the dystrophin gene. Sequencing is now ongoing to search for point mutations. Our findings in this family of Labrador Retriever dogs lend support to the hypothesis that, in exceptional situations, muscle with no dystrophin may be functional. Unlocking the secrets that protect these dogs from a severe clinical myopathy is a great challenge which may have important implications for future treatment of human muscular dystrophies.


Assuntos
Doenças do Cão/metabolismo , Distrofina/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/metabolismo , Animais , Modelos Animais de Doenças , Doenças do Cão/etiologia , Doenças do Cão/patologia , Cães , Família , Masculino , Músculo Esquelético/patologia , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/etiologia , Linhagem , Fenótipo , Utrofina/metabolismo
7.
Free Radic Biol Med ; 83: 129-38, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25668720

RESUMO

Nutritional muscular dystrophy (NMD) of chicks is induced by dietary selenium (Se)/vitamin E (Vit. E) deficiencies and may be associated with oxidative cell damage. To reveal the underlying mechanisms related to the presumed oxidative cell damage, we fed four groups of 1-day-old broiler chicks (n = 40/group) with a basal diet (BD; 10 µg Se/kg; no Vit. E added, -Se -Vit. E) or the BD plus all-rac-α-tocopheryl acetate at 50mg/kg (-Se +Vit. E), Se (as sodium selenite) at 0.3mg/kg (+Se -Vit. E), or both of these nutrients (+Se +Vit. E) for 6 weeks. High incidences of NMD (93%) and mortality (36%) of the chicks were induced by the BD, starting at week 3. Dietary Se deficiency alone also induced muscle fiber rupture and coagulation necrosis in the pectoral muscle of chicks at week 3 and thereafter, with increased (P < 0.05) malondialdehyde, decreased (P < 0.05) total antioxidant capacity, and diminished (P < 0.05) glutathione peroxidase activities in the muscle. To link these oxidative damages of the muscle cells to the Se-deficiency-induced NMD, we first determined gene expression of the potential 26 selenoproteins in the muscle of the chicks at week 2 before the onset of symptoms. Compared with the +Se chicks, the -Se chicks had lower (P < 0.05) muscle mRNA levels of Gpx1, Gpx3, Gpx4, Sepp1, Selo, Selk, Selu, Selh, Selm, Sepw1, and Sep15. The -Se chicks also had decreased (P < 0.05) production of 6 selenoproteins (long-form selenoprotein P (SelP-L), GPx1, GPx4, Sep15, SelW, and SelN), but increased levels (P < 0.05) of the short-form selenoprotein P in muscle at weeks 2 and 4. Dietary Se deficiency elevated (P < 0.05) muscle p53, cleaved caspase 3, cleaved caspase 9, cyclooxygenase 2 (COX2), focal adhesion kinase (FAK), phosphatidylinositol 3-kinase (PI3K), phospho-Akt, nuclear factor-κB (NF-κB), p38 mitogen-activated protein kinase (p38 MAPK), phospho-p38 MAPK, phospho-JNK, and phospho-ERK and decreased (P < 0.05) muscle procaspase 3, procaspase 9, and NF-κB inhibitor α. In conclusion, the downregulation of SelP-L, GPx1, GPx4, Sep15, SelW, and SelN by dietary Se deficiency might account for induced oxidative stress and the subsequent peroxidative damage of chick muscle cells via the activation of the p53/caspase 9/caspase 3, COX2/FAK/PI3K/Akt/NF-κB, and p38 MAPK/JNK/ERK signaling pathways. Metabolism of peroxides and redox regulation are likely to be the mechanisms whereby these selenoproteins prevented the onset of NMD in chicks.


Assuntos
Apoptose , Dieta/efeitos adversos , Distrofia Muscular Animal/prevenção & controle , Peróxidos/metabolismo , Selenoproteínas/metabolismo , Animais , Antioxidantes , Western Blotting , Proliferação de Células , Células Cultivadas , Galinhas , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Técnicas Imunoenzimáticas , Masculino , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/metabolismo , Oxirredução , Estresse Oxidativo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Selênio/metabolismo , Selenoproteínas/genética , Glutationa Peroxidase GPX1
8.
Int Rev Cell Mol Biol ; 300: 51-83, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23273859

RESUMO

Embryonic and postnatal development of skeletal muscle entails highly regulated processes whose complexity continues to be deconstructed. One key stage of development is the satellite cell, whose niche is composed of multiple cell types that eventually contribute to terminally differentiated myotubes. Understanding these developmental processes will ultimately facilitate treatments of myopathies such as Duchenne muscular dystrophy (DMD), a disease characterized by compromised cell membrane structure, resulting in severe muscle wasting. One theoretical approach is to use pluripotent stem cells in a therapeutic setting to help replace degenerated muscle tissue. This chapter discusses key myogenic developmental stages and their regulatory pathways; artificial myogenic induction in pluripotent stem cells; advantages and disadvantages of DMD animal models; and therapeutic approaches targeting DMD. Furthermore, skeletal muscle serves as an excellent paradigm for understanding general cell fate decisions throughout development.


Assuntos
Desenvolvimento Muscular/fisiologia , Músculo Esquelético/crescimento & desenvolvimento , Músculo Esquelético/fisiologia , Regeneração/fisiologia , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Cães , Corpos Embrioides/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , MicroRNAs/genética , Desenvolvimento Muscular/genética , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/terapia , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/fisiologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante , Receptores Notch/fisiologia , Regeneração/genética , Medicina Regenerativa/métodos , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/fisiologia , Transdução de Sinais , Nicho de Células-Tronco , Via de Sinalização Wnt
9.
Dev Biol ; 352(2): 228-42, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21256839

RESUMO

In Drosophila, like in humans, Dystrophin Glycoprotein Complex (DGC) deficiencies cause a life span shortening disease, associated with muscle dysfunction. We performed the first in vivo genetic interaction screen in ageing dystrophic muscles and identified genes that have not been shown before to have a role in the development of muscular dystrophy and interact with dystrophin and/or dystroglycan. Mutations in many of the found interacting genes cause age-dependent morphological and heat-induced physiological defects in muscles, suggesting their importance in the tissue. Majority of them is phylogenetically conserved and implicated in human disorders, mainly tumors and myopathies. Functionally they can be divided into three main categories: proteins involved in communication between muscle and neuron, and interestingly, in mechanical and cellular stress response pathways. Our data show that stress induces muscle degeneration and accelerates age-dependent muscular dystrophy. Dystrophic muscles are already compromised; and as a consequence they are less adaptive and more sensitive to energetic stress and to changes in the ambient temperature. However, only dystroglycan, but not dystrophin deficiency causes extreme myodegeneration induced by energetic stress suggesting that dystroglycan might be a component of the low-energy pathway and act as a transducer of energetic stress in normal and dystrophic muscles.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila/genética , Drosophila/metabolismo , Distroglicanas/genética , Distroglicanas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/metabolismo , Estresse Fisiológico , Animais , Sequência de Bases , Primers do DNA/genética , Modelos Animais de Doenças , Distroglicanas/antagonistas & inibidores , Distroglicanas/deficiência , Distrofina/antagonistas & inibidores , Distrofina/deficiência , Feminino , Genes de Insetos , Humanos , Masculino , Células Musculares/metabolismo , Distrofia Muscular Animal/etiologia , Mutação , Interferência de RNA , Transdução de Sinais
10.
Proc Natl Acad Sci U S A ; 106(31): 12573-9, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19633189

RESUMO

Skeletal muscle basal lamina is linked to the sarcolemma through transmembrane receptors, including integrins and dystroglycan. The function of dystroglycan relies critically on posttranslational glycosylation, a common target shared by a genetically heterogeneous group of muscular dystrophies characterized by alpha-dystroglycan hypoglycosylation. Here we show that both dystroglycan and integrin alpha7 contribute to force-production of muscles, but that only disruption of dystroglycan causes detachment of the basal lamina from the sarcolemma and renders muscle prone to contraction-induced injury. These phenotypes of dystroglycan-null muscles are recapitulated by Large(myd) muscles, which have an intact dystrophin-glycoprotein complex and lack only the laminin globular domain-binding motif on alpha-dystroglycan. Compromised sarcolemmal integrity is directly shown in Large(myd) muscles and similarly in normal muscles when arenaviruses compete with matrix proteins for binding alpha-dystroglycan. These data provide direct mechanistic insight into how the dystroglycan-linked basal lamina contributes to the maintenance of sarcolemmal integrity and protects muscles from damage.


Assuntos
Membrana Basal/fisiologia , Distroglicanas/fisiologia , Laminina/fisiologia , Sarcolema/fisiologia , Animais , Sítios de Ligação , Distroglicanas/química , Glicosilação , Integrinas/fisiologia , Laminina/química , Vírus da Coriomeningite Linfocítica , Camundongos , Distrofia Muscular Animal/etiologia
11.
Dev Dyn ; 238(8): 1878-86, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19582871

RESUMO

EMAP-like proteins (ELPs) are conserved microtubule-binding proteins that function during cell division and in the behavior of post-mitotic cells. In Caenorhabditis elegans, ELP-1 is broadly expressed in many cells and tissues including the touch receptor neurons and body wall muscle. Within muscle, ELP-1 is associated with a microtubule network that is closely opposed to the integrin-based adhesion sites called dense bodies. To examine ELP-1 function, we utilized an elp-1 RNA interference assay and screened for synthetic interactions with mutated adhesion site proteins. We reveal a synthetic lethal relationship between ELP-1 and the dystrophin-like protein, DYS-1. Reduction of ELP-1 in a dystrophin [dys-1(cx18)] mutant results in adult animals with motility defects, splayed and hypercontracted muscle with altered cholinergic signaling. Worms fill with vesicles, become flaccid, and die. We conclude that ELP-1 is a genetic modifier of a C. elegans model of muscular dystrophy.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/fisiologia , Distrofina/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Distrofia Muscular Animal/etiologia , Acetilcolina/fisiologia , Animais , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Distrofina/deficiência , Distrofina/genética , Genes de Helmintos , Genes Letais , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Contração Muscular/fisiologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Fenótipo , Interferência de RNA , Transdução de Sinais/fisiologia , Temperatura
12.
Neurobiol Dis ; 32(2): 243-53, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18694830

RESUMO

Aminoglycosides force read through of premature stop codon mutations and introduce new mutation-specific gene-corrective strategies in Duchenne muscular dystrophy. A chronic treatment with gentamicin (32 mg/kg/daily i.p., 8-12 weeks) was performed in exercised mdx mice with the dual aim to clarify the dependence on dystrophin of the functional, biochemical and histological alterations present in dystrophic muscle and to verify the long term efficiency of small molecule gene-corrective strategies in work-loaded dystrophic muscle. The treatment counteracted the exercise-induced impairment of in vivo forelimb strength after 6-8 weeks. We observed an increase in dystrophin expression level in all the fibers, although lower than that observed in normal fibers, and found a concomitant recovery of aquaporin-4 at sarcolemma. A significant reduction in centronucleated fibers, in the area of necrosis and in the percentage of nuclear factor-kB-positive nuclei was observed in gastrocnemious muscle of treated animals. Plasma creatine kinase was reduced by 70%. Ex vivo, gentamicin restored membrane ionic conductance in mdx diaphragm and limb muscle fibers. No effects were observed on the altered calcium homeostasis and sarcolemmal calcium permeability, detected by electrophysiological and microspectrofluorimetric approaches. Thus, the maintenance of a partial level of dystrophin is sufficient to reinforce sarcolemmal stability, reducing leakiness, inflammation and fiber damage, while correction of altered calcium homeostasis needs greater expression of dystrophin or direct interventions on the channels involved.


Assuntos
Distrofina/metabolismo , Gentamicinas/uso terapêutico , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal , Inibidores da Síntese de Proteínas/uso terapêutico , Animais , Aquaporina 4/metabolismo , Peso Corporal/efeitos dos fármacos , Cálcio/metabolismo , Homeostase/efeitos dos fármacos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos mdx , Força Muscular/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/patologia , Técnicas de Patch-Clamp , Condicionamento Físico Animal/efeitos adversos , Sarcolema/efeitos dos fármacos , Sarcolema/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
13.
J Biomed Sci ; 15(5): 595-604, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18459070

RESUMO

Myostatin, a member of the TGF-beta superfamily, is a potent negative regulator of skeletal muscle and growth. Previously, we reported Mstn1 from zebrafish and studied its influence on muscle development. In this study, we identified another form of Myostatin protein which is referred to as Mstn2. The size of Mstn2 cDNA is 1342 bp with 109 and 132 bp of 5' and 3'-untranslated regions (UTRs), respectively. The coding region is 1101 bp encoding 367 amino acids. The identity between zebrafish Mstn1 and 2 is 66%. The phylogenetic tree revealed that the Mstn2 is an ancestral form of Mstn1. To study the functional aspects, we overexpressed mstn2 and noticed that embryos became less active and the juveniles with bent and curved phenotypes when compared to the control. The RT-PCR and in situ hybridization showed concurrent reduction of dystrophin associated protein complex (DAPC). In cryosection and in situ hybridization, we observed the disintegration of somites, lack of transverse myoseptum and loss of muscle integrity due to the failure of muscle attachment in mstn2 overexpressed embryos. Immunohistochemistry and western blot showed that there was a reduction of dystrophin, dystroglycan and sarcoglycan at translational level in overexpressed embryos. Taken together, these results indicate the suitability of zebrafish as an excellent animal model and our data provide the first in vivo evidence of muscle attachment failure by the overexpression of mstn2 and it leads to muscle loss which results in muscle dystrophy that may contribute to Duchenne syndrome and other muscle related diseases.


Assuntos
Regulação para Baixo/genética , Complexo de Proteínas Associadas Distrofina/genética , Distrofina/genética , Distrofia Muscular Animal/etiologia , Miostatina/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Distroglicanas/genética , Embrião não Mamífero , Músculo Esquelético/fisiopatologia , Miostatina/genética , Fenótipo , Sarcoglicanas/genética , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
15.
J Physiol ; 586(7): 2003-14, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18258657

RESUMO

Duchenne muscular dystrophy (DMD) is a severe degenerative muscle disease caused by a mutation in the gene encoding dystrophin, a protein linking the cytoskeleton to the extracellular matrix. In this study we investigated whether the antioxidant N-acetylcysteine (NAC) provided protection against dystrophic muscle damage in the mdx mouse, an animal model of DMD. In isolated mdx muscles, NAC prevented the increased membrane permeability and reduced the force deficit associated with stretch-induced muscle damage. Three-week-old mdx mice were treated with NAC in the drinking water for 6 weeks. Dihydroethidium staining showed that NAC treatment reduced the concentration of reactive oxygen species (ROS) in mdx muscles. This was accompanied by a significant decrease in centrally nucleated fibres in muscles from NAC-treated mdx mice. Immunoblotting showed that NAC treatment decreased the nuclear protein expression of NF-kappaB, a transcription factor involved in pro-inflammatory cytokine expression. Finally, we show that NAC treatment reduced caveolin-3 protein levels and increased the sarcolemmal expression of beta-dystroglycan and the dystrophin homologue, utrophin. Taken together, our findings suggest that ROS play an important role in the dystrophic pathogenesis, both in terms of activating damage pathways and in regulating the expression of some dystrophin-associated membrane proteins. These results offer the prospect that antioxidants such as NAC could have therapeutic potential for DMD patients.


Assuntos
Acetilcisteína/uso terapêutico , Sequestradores de Radicais Livres/uso terapêutico , Músculo Esquelético/fisiopatologia , Distrofia Muscular Animal/prevenção & controle , Acetilcisteína/farmacologia , Animais , Caveolina 3/metabolismo , Modelos Animais de Doenças , Distroglicanas/metabolismo , Distrofina/genética , Distrofina/metabolismo , Sequestradores de Radicais Livres/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/fisiopatologia , Distrofia Muscular de Duchenne/fisiopatologia , Distrofia Muscular de Duchenne/prevenção & controle , Mutação/genética , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo
16.
Aviakosm Ekolog Med ; 42(4): 31-5, 2008.
Artigo em Russo | MEDLINE | ID: mdl-19140470

RESUMO

The neuromuscular junctions were investigated in m. soleus of rats subjected to a 3-month tail suspension simulating the microgravity effects. Electron microscopy analysis revealed some ultrastructural signs of atrophy, degeneration and adipose dystrophy of muscle fibers. The aggregate of these findings points to progressive atrophy in m. soleus, while ultrastructural changes in the neuromuscular synapses testify a reduced functional activity of the synapses and partial denervation of the muscle fibers which, probably, underlay the atrophic process in the muscle. Increases in the number of axon terminals found in some neuromuscular synapses as well as of synaptic vesicles in individual axon terminals are likely to reflect formation of a particularly active pool of spinal motoneurons at L5, possibly associated with the growth in the number of fast fibers resulted from transformation.


Assuntos
Neurônios Motores/ultraestrutura , Músculo Esquelético/inervação , Junção Neuromuscular/ultraestrutura , Simulação de Ausência de Peso/efeitos adversos , Animais , Modelos Animais de Doenças , Progressão da Doença , Seguimentos , Elevação dos Membros Posteriores/efeitos adversos , Elevação dos Membros Posteriores/métodos , Masculino , Microscopia Eletrônica , Músculo Esquelético/ultraestrutura , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/patologia , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Wistar , Vesículas Sinápticas/ultraestrutura , Fatores de Tempo , Simulação de Ausência de Peso/veterinária
17.
PLoS One ; 2(8): e806, 2007 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-17726536

RESUMO

BACKGROUND: Duchenne Muscular Dystrophy (DMD) is characterized by increased muscle damage and an abnormal blood flow after muscle contraction: the state of functional ischemia. Until now, however, the cause-effect relationship between the pathogenesis of DMD and functional ischemia was unclear. We examined (i) whether functional ischemia is necessary to cause contraction-induced myofiber damage and (ii) whether functional ischemia alone is sufficient to induce the damage. METHODOLOGY/PRINCIPAL FINDINGS: In vivo microscopy was used to document assays developed to measure intramuscular red blood cell flux, to quantify the amount of vasodilatory molecules produced from myofibers, and to determine the extent of myofiber damage. Reversal of functional ischemia via pharmacological manipulation prevented contraction-induced myofiber damage in mdx mice, the murine equivalent of DMD. This result indicates that functional ischemia is required for, and thus an essential cause of, muscle damage in mdx mice. Next, to determine whether functional ischemia alone is enough to explain the disease, the extent of ischemia and the amount of myofiber damage were compared both in control and mdx mice. In control mice, functional ischemia alone was found insufficient to cause a similar degree of myofiber damage observed in mdx mice. Additional mechanisms are likely contributing to cause more severe myofiber damage in mdx mice, suggestive of the existence of a "two-hit" mechanism in the pathogenesis of this disease. CONCLUSIONS/SIGNIFICANCE: Evidence was provided supporting the essential role of functional ischemia in contraction-induced myofiber damage in mdx mice. Furthermore, the first quantitative evidence for the "two-hit" mechanism in this disease was documented. Significantly, the vasoactive drug tadalafil, a phosphodiesterase 5 inhibitor, administered to mdx mice ameliorated muscle damage.


Assuntos
Isquemia/complicações , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/etiologia , Inibidores da Fosfodiesterase 5 , Animais , Carbolinas/uso terapêutico , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Modelos Animais de Doenças , Peróxido de Hidrogênio/metabolismo , Camundongos , Camundongos Transgênicos , Contração Muscular , Distrofia Muscular Animal/enzimologia , Óxido Nítrico/biossíntese , Inibidores de Fosfodiesterase/uso terapêutico , Tadalafila
18.
EMBO J ; 26(2): 481-93, 2007 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-17215867

RESUMO

Perturbation in the Dystroglycan (Dg)-Dystrophin (Dys) complex results in muscular dystrophies and brain abnormalities in human. Here we report that Drosophila is an excellent genetically tractable model to study muscular dystrophies and neuronal abnormalities caused by defects in this complex. Using a fluorescence polarization assay, we show a high conservation in Dg-Dys interaction between human and Drosophila. Genetic and RNAi-induced perturbations of Dg and Dys in Drosophila cause cell polarity and muscular dystrophy phenotypes: decreased mobility, age-dependent muscle degeneration and defective photoreceptor path-finding. Dg and Dys are required in targeting glial cells and neurons for correct neuronal migration. Importantly, we now report that Dg interacts with insulin receptor and Nck/Dock SH2/SH3-adaptor molecule in photoreceptor path-finding. This is the first demonstration of a genetic interaction between Dg and InR.


Assuntos
Modelos Animais de Doenças , Drosophila , Distroglicanas/metabolismo , Distrofina/metabolismo , Distrofia Muscular Animal/genética , Malformações do Sistema Nervoso/genética , Proteínas Adaptadoras de Transdução de Sinal , Animais , Animais Geneticamente Modificados , Movimento Celular/genética , Polaridade Celular , Distroglicanas/genética , Distroglicanas/fisiologia , Distrofina/genética , Distrofina/fisiologia , Humanos , Modelos Biológicos , Modelos Moleculares , Distrofia Muscular Animal/etiologia , Mutação , Proteínas Oncogênicas/metabolismo
19.
Braz. j. morphol. sci ; 23(2): 217-222, Apr.-June 2006. ilus
Artigo em Inglês | LILACS | ID: lil-468068

RESUMO

The murine model of muscular dystrophy, the mdx mice, is widely used to study the pathogenesis of muscular dystrophies. These mice suffer an X-linked dystrophin deficiency and present cycles of muscle fiber degeneration-regeneration beginning at 21 days of age. At the present, we studied neuromuscular junction organization in the sternomastoid muscle of mdx mice, focusing on the distribution of terminal Schwann cells during early development and adults. Seven and 14 days after birth (n=200 endplates for each age), before the onset of muscle degeneration-regeneration, fluorescence confocal microscopy showed that there were no detectable differences in the pattern of Schwann cell distribution in the mdx compared to controls of the same age. Schwann cells had a diffuse pattern of distribution, covering the plaques of acetylcholine receptors. In adult mdx muscles, terminal Schwann cell processes filled the center of acetylcholine receptors islands, similar to nerve terminal distribution, at the majority of the junctions (n=200; 100%). Conversely, all of the adult control junctions (n=200) showed continuous processes of Schwann cells covering the continuous branches of acetylcholine receptors. These observations indicate that remodeling of the three components of the neuromuscular junction occurs only after the onset of the cycles of muscle fiber degeneration-regeneration, in the mdx mice.


Assuntos
Animais , Camundongos , Células de Schwann/citologia , Células de Schwann/fisiologia , Distrofia Muscular Animal/etiologia , Junção Neuromuscular/fisiologia , Células de Schwann , Transmissão Sináptica , Camundongos Endogâmicos mdx , Junção Neuromuscular
20.
Trends Cell Biol ; 16(4): 198-205, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16515861

RESUMO

The dystrophin-glycoprotein complex (DGC) can be considered as a specialized adhesion complex, linking the extracellular matrix to the actin cytoskeleton, primarily in muscle cells. Mutations in several components of the DGC lead to its partial or total loss, resulting in various forms of muscular dystrophy. These typically manifest as progressive wasting diseases with loss of muscle integrity. Debate is ongoing about the precise function of the DGC: initially a strictly mechanical role was proposed but it has been suggested that there is aberrant calcium handling in muscular dystrophy and, more recently, changes in MAP kinase and GTPase signalling have been implicated in the aetiology of the disease. Here, we discuss new and interesting developments in these aspects of DGC function and attempt to rationalize the mechanical, calcium and signalling hypotheses to provide a unifying hypothesis of the underlying process of muscular dystrophy.


Assuntos
Complexo de Proteínas Associadas Distrofina/fisiologia , Distrofina/fisiologia , Distrofia Muscular Animal/etiologia , Distrofia Muscular Animal/metabolismo , Transdução de Sinais , Animais , Cálcio/metabolismo , Moléculas de Adesão Celular/metabolismo , Citoesqueleto/metabolismo , Distrofina/genética , Complexo de Proteínas Associadas Distrofina/genética , Previsões , Sistema de Sinalização das MAP Quinases , Camundongos , Modelos Biológicos , Contração Muscular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA