Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Respir Crit Care Med ; 186(11): 1140-9, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23024021

RESUMO

RATIONALE: Mechanical ventilation (MV) is associated with adverse effects on the diaphragm, but the cellular basis for this phenomenon, referred to as ventilator-induced diaphragmatic dysfunction (VIDD), is poorly understood. OBJECTIVES: To determine whether mitochondrial function and cellular energy status are disrupted in human diaphragms after MV, and the role of mitochondria-derived oxidative stress in the development of VIDD. METHODS: Diaphragm and biceps specimens obtained from brain-dead organ donors who underwent MV (15-176 h) and age-matched control subjects were compared regarding mitochondrial enzymatic function, mitochondrial DNA integrity, lipid content, and metabolic gene and protein expression. In addition, diaphragmatic force and oxidative stress after exposure to MV for 6 hours were evaluated in mice under different conditions. MEASUREMENTS AND MAIN RESULTS: In human MV diaphragms, mitochondrial biogenesis and content were down-regulated, with a more specific defect of respiratory chain cytochrome-c oxidase. Laser capture microdissection of cytochrome-c oxidase-deficient fibers revealed mitochondrial DNA deletions, consistent with damage from oxidative stress. Diaphragmatic lipid accumulation and responses of master cellular metabolic sensors (AMP-activated protein kinase and sirtuins) were consistent with energy substrate excess as a possible stimulus for these changes. In mice, induction of hyperlipidemia worsened diaphragmatic oxidative stress during MV, whereas transgenic overexpression of a mitochondria-localized antioxidant (peroxiredoxin-3) was protective against VIDD. CONCLUSIONS: Our data suggest that mitochondrial dysfunction lies at the nexus between oxidative stress and the impaired diaphragmatic contractility that develops during MV. Energy substrate oversupply relative to demand, resulting from diaphragmatic inactivity during MV, could play an important role in this process.


Assuntos
Diafragma/metabolismo , Metabolismo dos Lipídeos/fisiologia , Mitofagia , Estresse Oxidativo/fisiologia , Respiração Artificial/efeitos adversos , Animais , Estudos de Casos e Controles , Diafragma/patologia , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Reação em Cadeia da Polimerase em Tempo Real , Valores de Referência , Estatísticas não Paramétricas , Técnicas de Cultura de Tecidos
2.
J Gene Med ; 12(3): 266-75, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20082422

RESUMO

BACKGROUND: Efficient adenovirus (AdV)-mediated gene transfer is possible only in immature muscle or regenerating muscle, suggesting that a developmentally regulated event plays a major role in limiting AdV uptake in mature skeletal muscle. Previously, we showed that the expression of the primary coxsackie and adenovirus receptor (CAR) is severely down-regulated during muscle maturation and that, in muscle-specific CAR transgenic mice, there is significant enhancement of AdV-mediated gene transfer to mature skeletal muscle. METHODS: To evaluate whether increasing CAR expression can also augment gene transfer to dystrophic muscle that has many regenerating fibers, we crossed CAR transgenics with dystrophin-deficient mice (mdx/CAR). We also tested a two-step protocol in which CAR levels were increased in the target muscle, prior to administration of AdV, through the use of recombinant adeno-associated virus (AAV2) expressing CAR. Lastly, we assessed the effect of histone deacetylase inhibitors on CAR and AdV transduction efficiency in myoblasts and mdx muscle. RESULTS: Although somewhat higher rates of transduction can be achieved in adult mdx mice than in normal mice as a result of ongoing muscle regeneration in these animals, CAR expression in the mdx background (mdx/CAR transgenics) still markedly improved the susceptibility of mature muscle to AdV-mediated gene transfer of dystrophin. Prior administration of AAV2-CAR to normal muscle led to significantly increased transduction by subsequent injection of AdV. The histone deacetylase inhibitor valproate increased CAR transcript and protein levels in myoblasts and mdx muscle, and also increased AdV-mediated gene transfer. CONCLUSIONS: We have developed a method of increasing CAR levels in both normal and regenerating muscle.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/genética , Receptores Virais/genética , Transdução Genética/métodos , Adenoviridae , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Distrofina/genética , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Regeneração , Transcrição Gênica/efeitos dos fármacos , Ácido Valproico/farmacologia
3.
Ann Neurol ; 65(1): 7-11, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19194875

RESUMO

Sporadic inclusion body myositis is the commonest acquired disease of skeletal muscles after 50 years of age, and as such it has commanded a great deal of attention of investigators over the past 25 years. As a result, a large amount of information has accumulated concerning its clinical profile, myopathology, and immunopathology. In the myopathology and immunopathology, there is general agreement that the characteristic features could be divided into a degenerative and an inflammatory group. However, there has been controversy about the possible role of these changes in the pathogenesis of muscle fiber damage. In particular, there is no agreement whether a cause-and-effect relationship exists between these two groups of changes, and if so, which is the primary one. In this brief overview, we examine the validity of the various controversial observations and critically review the justification for the two major hypotheses for the primary role of inflammation versus degeneration.


Assuntos
Miosite de Corpos de Inclusão/etiologia , Miosite de Corpos de Inclusão/patologia , Fatores Etários , Progressão da Doença , Humanos , Inflamação , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Miosite de Corpos de Inclusão/complicações
4.
J Muscle Res Cell Motil ; 31(3): 181-93, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20706865

RESUMO

Duchenne muscular dystrophy is a genetic muscle disease characterized by the absence of sub-sarcolemmal dystrophin that results in muscle fibre necrosis, progressive muscle wasting and is fatal. Numerous experimental studies with dystrophin-deficient mdx mice, an animal model for the disease, have demonstrated that extrasynaptic upregulation of utrophin, an analogue of dystrophin, can prevent muscle fibre deterioration and reduce or negate the dystrophic phenotype. A different approach for ectopic expression of utrophin relies on augmentation of CT-GalNAc transferase in muscle fibre. We investigated whether CT-GalNAc transferase overexpression in adult mice influence appearance of utrophin in the extrasynaptic sarcolemma. After electrotransfer of plasmid DNA carrying an expression cassette of CT-GalNAc transferase into tibialis anterior muscle of wild type and dystrophic mice, muscle sections were examined by immunofluorescence. CT-GalNAc transgene expression augmented sarcolemmal carbohydrate glycosylation and was accompanied by extrasynaptic utrophin. A 6-week time course study showed that the highest efficiency of utrophin overexpression in a plasmid harboured muscle fibres was 32.2% in CD-1 and 52% in mdx mice, 2 and 4 weeks after CT-GalNAc gene transfer, respectively. The study provides evidence that postnatal CT-GalNAc transferase overexpression stimulates utrophin upregulation that is inherently beneficial for muscle structure and strength restoration. Thus CT-GalNAc may provide an important therapeutic molecule for treatment of dystrophin deficiency in Duchenne muscular dystrophy.


Assuntos
Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , N-Acetilgalactosaminiltransferases/biossíntese , Junção Neuromuscular/metabolismo , Utrofina/metabolismo , Animais , Linhagem Celular , Modelos Animais de Doenças , Expressão Gênica , Glicosilação , Humanos , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/genética , N-Acetilgalactosaminiltransferases/genética , Junção Neuromuscular/genética , Sarcolema/genética , Sarcolema/metabolismo , Utrofina/genética
5.
Hum Gene Ther ; 19(2): 133-42, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18067405

RESUMO

Adenoviral vectors that use the coxsackievirus and adenovirus receptor do not transduce mature muscle efficiently. Group B adenoviruses use CD46 as their cell attachment receptor. To evaluate the utility of vectors based on group B adenoviruses for gene transfer to human skeletal muscle, we assessed the expression of CD46 in biopsied normal skeletal muscle samples and in muscles from patients with Duchenne muscular dystrophy. Transcript levels of CD46 were extremely low in mature muscle and CD46 immunoreactivity was detected only on blood vessels in the muscle sections. Although myoblasts cultured from biopsied samples had robust cell surface CD46 expression by flow cytometry, CD46 transcript levels were barely detectable after differentiation of the myoblasts into myotubes. The myotubes were also much less susceptible to infection with an adenoviral vector carrying the fiber of serotype 35 adenovirus (AdF35). These results suggest that for skeletal muscle, vectors derived from group B adenoviruses may not be a suitable alternative to the commonly used Ad5 vectors.


Assuntos
Adenoviridae/metabolismo , Diferenciação Celular , Regulação para Baixo/genética , Proteína Cofatora de Membrana/genética , Músculo Esquelético/citologia , Músculo Esquelético/metabolismo , Transdução Genética/métodos , Biópsia , Permeabilidade da Membrana Celular , Células Cultivadas , Citometria de Fluxo , Humanos , Fibras Musculares Esqueléticas , Distrofia Muscular de Duchenne , Mioblastos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta-Galactosidase
6.
Cell Transplant ; 17(3): 325-35, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18522235

RESUMO

Possible myogenic differentiation of SSEA-1- and OCT-4-positive murine embryonic stem cells (ESCs) and embryoid bodies (EBs) was studied in vitro and in vivo. In vitro, ESC- or EB-derived ESCs (EBs/ESCs) showed only traces of Pax 3 and 7 expression by immunocytochemistry and Pax 3 expression by immunoblot. By RT-PCR, myogenic determinant molecules (myf5, myoD, and myogenin) were expressed by EBs/ESCs but not by ESCs. However, in such cultures, very rare contracting myotubes were still present. Suspensions of LacZ-labeled ESCs or EBs were injected into anterior tibialis muscles (ATM) of different cohorts of mice for the study of their survival and possible myogenic differentiation. The different cohorts of mice included isogenic adult 129/Sv, nonisogenic CD1 and mdx, as well as mdx immunosuppressed with 2.5 mg/kg daily injections of tacrolimus. Ten to 90 days postinjections, the injected ATM of nonisogenic mice did not contain cells positive for LacZ, SSEA-1, OCT-4, or embryonic myosin heavy chain. The ATM of intact mdx mice contained very rare examples of muscle fibers positive for dystrophin and/or embryonic myosin heavy chain. In the ATM of the isogenic normal and the immunosuppressed mdx mice, as expected, large teratomas developed containing the usual diverse cell types. In some teratomas of immunosuppressed mdx mice, small pockets of muscle fibers expressed dystrophin and myosin heavy chain. Our studies indicated that in muscles of animals nonisogenic with the used ESCs, only very rare ESCs survived with myogenic differentiation. These studies also indicated that ESCs will not undergo significant, selective, and preferential myogenic differentiation in vitro or in vivo in any of the models studied. It is probable that this strain of murine ESC requires some experimentally induced alteration of its gene expression profile to secure significant myogenicity and suppress tumorogenicity.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Músculo Esquelético/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Distrofina/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Immunoblotting , Imuno-Histoquímica , Imunossupressores/farmacologia , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Proteína MyoD/genética , Fator Regulador Miogênico 5/genética , Miogenina/genética , Fator de Transcrição PAX3 , Fator de Transcrição PAX7/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tacrolimo/farmacologia
7.
J Neurol Sci ; 250(1-2): 71-8, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16996541

RESUMO

Dysferlin is a large sarcolemmal protein implicated in the repair of surface membrane tears in muscle cells. Mutations in dysferlin result in limb girdle muscular dystrophy type 2B and Miyoshi myopathy. Using a cDNA based approach we identified eight new pathogenic dysferlin alleles. To better understand how missense mutations could lead to reduced or absent dysferlin expression levels, we mapped missense mutations from our own and from published databases (n=55) to the secondary protein structure of dysferlin, deduced by computerized structural prediction tools. We found the protein to be very sensitive to the alteration of residues that were predicted to be buried inside the protein structure. We identified seven putative C2 domains, one more than commonly reported, of both type I and type II topology in dysferlin. Missense mutations often affected those structures as well as residues that were highly conserved between members of the ferlin family. Thus, alteration of structurally important residues in dysferlin could lead to improper folding and degradation of the mutant protein.


Assuntos
Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas Musculares/química , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Mutação de Sentido Incorreto/genética , Adolescente , Adulto , Sequência de Aminoácidos/genética , Criança , Sequência Conservada , Análise Mutacional de DNA , DNA Complementar/análise , Disferlina , Evolução Molecular , Predisposição Genética para Doença/genética , Humanos , Dados de Sequência Molecular , Músculo Esquelético/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Filogenia , Dobramento de Proteína , Estrutura Quaternária de Proteína/genética , Estrutura Terciária de Proteína/genética , Proteômica
8.
Can J Gastroenterol ; 20(6): 433-5, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16779462

RESUMO

The association between dermatomyositis and celiac disease in children has been well documented. In the adult population, however, the association has not been clearly established. A rare case of concomitant dermatomyositis and celiac disease in a 40-year-old woman is presented. After having been diagnosed with dermatomyositis and iron deficiency anemia, this patient was referred to the gastroenterology clinic to exclude a gastrointestinal malignancy. Blood tests revealed various vitamin deficiencies consistent with malabsorption. The results of gastroscopy with duodenal biopsy were consistent with celiac disease. After she was put on a strict gluten-free diet, both nutritional deficiencies and the dermatomyositis resolved. The patient's human leukocyte antigen haplotype study was positive for DR3 and DQ2, which have been shown to be associated with both juvenile dermatomyositis and celiac disease. It is suggested that patients with newly diagnosed dermatomyositis be investigated for concomitant celiac disease even in the absence of gastrointestinal symptoms.


Assuntos
Doença Celíaca/dietoterapia , Doença Celíaca/diagnóstico , Dermatomiosite/dietoterapia , Dermatomiosite/diagnóstico , Dieta com Restrição de Proteínas/métodos , Adulto , Doença Celíaca/complicações , Dermatomiosite/complicações , Feminino , Glutens/administração & dosagem , Antígenos HLA-DQ/análise , Antígeno HLA-DR3/análise , Humanos , Resultado do Tratamento
9.
Hum Gene Ther ; 16(4): 489-501, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15871680

RESUMO

Chronic inflammation in tibialis anterior muscles of mdx mice was produced by a single injection of a recombinant adenovirus vector (AV) expressing an immunogenic beta-galactosidase (beta-gal). In regions of intense beta-gal staining, mononuclear infiltrates abounded, and muscle fibers showed strong extrasynaptic utrophin immunostaining, restoration of dystrophin-associated protein complex, and a marked reduction of the prevalence of centronucleation. Immunoblot analysis confirmed an increase of endogenous utrophin without an increase of the mRNA of the major muscle isoform utrA. Significantly better maximal tetanic force values were demonstrated in the inflammatory versus control mdx muscles. The resistance to lengthening contraction- induced damage was also significantly increased in the former. In muscles of mice lacking TNF-alpha gene, AV vector did not induce inflammation and extrajunctional utrophin increase did not occur. In the inflammatory mdx muscles, proteolytic activity of calcium-activated calpain was reduced, and in mdx myotubes in vitro, incubation with NO donors also reduced calpain-mediated utrophin proteolysis. Since utrophin was shown to be a natural substrate of calpain and known inhibitors of calpain in cultured mdx myotubes increased utrophin levels, the above results were consistent with the following conclusions: (1) extrasynaptic utrophin increase is mainly responsible for the antidystrophic effect; (2) extrasynaptic utrophin increase is a result of posttranscriptional mechanism(s) related to proinflammatory factors; and (3) reduction of endogenous muscle calpain activity by inflammatory cytokines has an important role in the stabilization and increase of the extrasynaptic utrophin.


Assuntos
Miosite/metabolismo , Sarcolema/metabolismo , Utrofina/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Calpaína/metabolismo , Células Cultivadas , Doença Crônica , Citocinas/genética , Citocinas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos mdx , Camundongos Knockout , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/imunologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Miosite/etiologia , Miosite/patologia , Doadores de Óxido Nítrico/farmacologia , Processamento de Proteína Pós-Traducional , Sinapses/metabolismo , Utrofina/efeitos dos fármacos , Utrofina/genética , beta-Galactosidase/efeitos adversos , beta-Galactosidase/genética
10.
Hum Gene Ther ; 15(2): 145-56, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14975187

RESUMO

Dystrophin gene transfer using helper-dependent adenoviral vectors (HDAd) deleted of all viral genes is a promising option to treat muscles in Duchenne muscular dystrophy (DMD). Previously, we reported high-level dystrophin expression and functional correction of dystrophin-deficient (mdx) mouse muscle 60 days after gene transfer with an HDAd encoding two full-length murine dystrophin cDNAs (referred to as HDCBDysM). In the present study, we tested the long-term efficacy of HDCBDysM by examining muscle contractility parameters and the stability of dystrophin expression 1 year after injection into neonatal mdx muscles. At this point, HDCBDysM-treated muscles averaged 52% dystrophin-positive fibers. Treated muscles also displayed significantly greater isometric force production as well as greater resistance to the force deficits and damage caused by eccentric contractions. The level of protection against eccentric contraction-induced force deficits correlated with the percentage of dystrophin-positive fibers. Furthermore, HDCBDysM treatment restored the dystrophin-glycoprotein complex (DGC) to the sarcolemma and improved other aspects of mdx muscle histopathology examined (central nucleation, muscle hypertrophy, and mononuclear [phagocytic] cell infiltration). These improvements occurred despite the induction of a humoral response against murine dystrophin. Our results indicate that major therapeutic benefits of HDCBDysM are maintained for a long period of the animals' lifespan and suggest that HDCBDys holds promise for treating DMD by gene therapy.


Assuntos
Adenoviridae/genética , Distrofina/genética , Vetores Genéticos , Vírus Auxiliares/genética , Contração Muscular , Distrofia Muscular de Duchenne/terapia , Animais , Animais Recém-Nascidos , Proteínas do Citoesqueleto/metabolismo , Proteínas do Citoesqueleto/ultraestrutura , DNA/genética , DNA/metabolismo , Distroglicanas , Distrofina/metabolismo , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/ultraestrutura , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/imunologia , Distrofia Muscular de Duchenne/fisiopatologia , Fatores de Tempo , Transdução Genética
11.
J Neuropathol Exp Neurol ; 62(12): 1203-10, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14692696

RESUMO

This review covers, in general terms, the salient features and impact of molecular myology under the following headings: its role in providing clues for the understanding of molecular etiology and pathogenesis of genetic myopathies, its contribution to the modernization and rationalization of the classification of muscle diseases, providing means of precise diagnosis and prevention of myopathies, development of radically new cell and gene therapies, and determination of future research directions. Myology appears to be among the medical disciplines that have benefited a great deal from molecular science. This remarkable progress will hopefully translate into effective treatment capabilities in the near future.


Assuntos
Doenças Musculares , Animais , Terapia Genética/métodos , Terapia Genética/tendências , Humanos , Doenças Musculares/classificação , Doenças Musculares/diagnóstico , Doenças Musculares/genética , Doenças Musculares/terapia
12.
BMC Cell Biol ; 5(1): 42, 2004 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-15533241

RESUMO

BACKGROUND: The Coxsackie and adenovirus receptor (CAR) has a restricted expression pattern in the adult. In skeletal muscle, although CAR is expressed in immature fibers, its transcript levels are barely detectable in mature muscle. This is in contrast to the robust expression observed in the heart. However, both heart and skeletal muscle are susceptible to infection with the Coxsackie B virus which utilizes primarily CAR for cellular internalization. The specific point of viral entry in skeletal and heart muscle remains unknown. RESULTS: Using antibodies directed against the extracellular and the cytoplasmic domains of CAR, we show CAR in normal human and mouse skeletal muscle to be a novel component of the neuromuscular junction. In cardiac muscle, CAR immunoreactivity is observed at the level of intercalated discs. We demonstrate a single isoform of CAR to be expressed exclusively at the human neuromuscular junction whereas both predominant CAR isoforms are expressed at the intercalated discs of non-diseased human heart. CONCLUSION: The localization of CAR to these important junctional complexes suggests that CAR may play both a structural and a regulatory role in skeletal and cardiac muscle, and that these complexes may serve as a point of entry for Coxsackie B virus.


Assuntos
Junções Intercelulares/metabolismo , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Junção Neuromuscular/metabolismo , Receptores Virais/metabolismo , Sequência de Aminoácidos , Animais , Proteína de Membrana Semelhante a Receptor de Coxsackie e Adenovirus , Humanos , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Junção Neuromuscular/química , Isoformas de Proteínas/análise , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores Virais/análise , Receptores Virais/química
14.
Can J Neurol Sci ; 30(3): 244-51, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12945950

RESUMO

BACKGROUND: Oculopharyngeal muscular dystrophy (OPMD) is an adult-onset disorder characterized by progressive ptosis, dysphagia and proximal limb weakness. The autosomal dominant form of this disease is caused by short expansions of a (GCG)6 repeat to (GCG) in the PABPN1 gene. The mutations lead to the expansion of a polyalanine stretch from 10 to 12-17 alanines in the N-terminus of PABPN1. The mutated PABPN1 (mPABPN1) induces the formation of intranuclear filamentous inclusions that sequester poly(A) RNA and are associated with cell death. METHODS: Human fetal brain cDNA library was used to look for PABPNI binding proteins using yeast two-hybrid screen. The protein interaction was confirmed by GST pull-down and co-immunoprecipitation assays. Oculopharyngeal muscular dystrophy cellular model and OPMD patient muscle tissue were used to check whether the PABPN1 binding proteins were involved in the formation of OPMD intranuclear inclusions. RESULTS: We identify two PABPNI interacting proteins, hnRNP A1 and hnRNP A/B. When co-expressed with mPABPN1 in COS-7 cells, predominantly nuclear protein hnRNP A1 and A/B co-localize with mPABPN1 in the insoluble intranuclear aggregates. Patient studies showed that hnRNP A1 is sequestered in OPMD nuclear inclusions. CONCLUSIONS: The hnRNP proteins are involved in mRNA processing and mRNA nucleocytoplasmic export, sequestering of hnRNPs in OPMD intranuclear aggregates supports the view that OPMD intranuclear inclusions are "poly(A) RNA traps", which would interfere with RNA export, and cause muscle cell death.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Distrofia Muscular Oculofaríngea/metabolismo , Proteína II de Ligação a Poli(A)/metabolismo , Animais , Células COS , Núcleo Celular/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1 , Humanos , Corpos de Inclusão/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Peptídeos/farmacologia , Proteína II de Ligação a Poli(A)/química , Proteína II de Ligação a Poli(A)/efeitos dos fármacos , Testes de Precipitina , Solubilidade , Distribuição Tecidual
15.
Hum Gene Ther ; 21(1): 127-34, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19719387

RESUMO

High-level tissue-specific expression of recombinant proteins in muscle is an important issue for several therapeutic applications. To achieve this goal, we generated several constructs containing one to five copies of the upstream enhancer (USE) of 160-bp of the human slow troponin I gene, linked to that gene's minimal promoter. We also tested constructs made with one to four copies of a 100-bp deletion of USE (DeltaUSE) reported to drive pan-muscle-specific expression in transgenic mice. These constructs were evaluated by measuring the activity of the reporter gene beta-galactosidase (beta-gal). In cell culture, these multimerized enhancers retained tissue specificity and their transcriptional strength increased with the number of enhancer copies. In myotube cultures (which still contain nondifferentiated cells), constructs containing four and five USE copies were stronger than the cytomegalovirus (CMV) early enhancer/promoter and slightly weaker than the hybrid CMV enhancer/beta-actin (CB) promoter. Those containing three USE, or four DeltaUSE copies were similar in strength to CMV. After electrotransfer of plasmid DNA into the mouse tibialis anterior muscle, the strengths of the two constructs (USEx3 and DeltaUSEx3) were tested; as measured by beta-gal activity in the total muscle lysate and by the number of transduced fibers, they were similar to CMV and CB. Muscle fiber typing, after electrotransfer of the soleus muscle, showed that DeltaUSEx3 and USEx3 were active in slow and fast fibers. The tissue specificity of these two constructs was also evaluated by hydrodynamic plasmid injection through the tail vein. Although significant beta-gal expression was measured in the liver when CMV was tested, no expression above background level was detected with USEx3 and DeltaUSEx3. The strength, muscle specificity, and small size of these transcriptional elements render them very attractive for gene therapy applications.


Assuntos
Elementos Facilitadores Genéticos/genética , Dosagem de Genes/genética , Regulação da Expressão Gênica/genética , Engenharia Genética , Músculo Esquelético/metabolismo , Troponina I/genética , Animais , Sequência de Bases , Células Cultivadas , Genes Reporter , Humanos , Camundongos , Fibras Musculares de Contração Rápida/citologia , Fibras Musculares de Contração Rápida/metabolismo , Fibras Musculares de Contração Lenta/citologia , Fibras Musculares de Contração Lenta/metabolismo , Músculo Esquelético/citologia , Especificidade de Órgãos/genética , Plasmídeos/genética , beta-Galactosidase/metabolismo
17.
Muscle Nerve ; 37(3): 338-42, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18004770

RESUMO

Danon disease is a rare X-linked dominant disorder caused by lysosomal-associated membrane protein 2 (LAMP2) deficiency and is characterized by hypertrophic cardiomyopathy, cardiac conduction abnormalities, skeletal vacuolar myopathy, variable degree of mental retardation, and peripheral pigmentary retinopathy. We describe a novel splice mutation in the LAMP2 gene in a French Canadian family. By identifying this novel mutation we were able to offer genetic screening and counseling to all family members. Presymptomatic diagnosis is important as cardiac surveillance can be life-saving.


Assuntos
Saúde da Família , Doença de Depósito de Glicogênio Tipo IIb/genética , Proteína 2 de Membrana Associada ao Lisossomo/genética , Mutação/genética , Adulto , Idoso , Análise Mutacional de DNA/métodos , Éxons/genética , Feminino , Doença de Depósito de Glicogênio Tipo IIb/patologia , Humanos , Masculino , Músculo Esquelético/patologia
18.
J Biol Chem ; 283(50): 34720-7, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18945675

RESUMO

Duchenne muscular dystrophy is caused by a genetic defect in the dystrophin gene. The absence of dystrophin results in muscle fiber necrosis and regeneration, leading to progressive muscle fiber loss. Utrophin is a close analogue of dystrophin. A substantial, ectopic expression of utrophin in the extrasynaptic sarcolemma of dystrophin-deficient muscle fibers can prevent deleterious effects of dystrophin deficiency. An alternative approach for the extrasynaptic up-regulation of utrophin involves the augmentation of utrophin transcription via the endogenous utrophin A promoter using custom-designed transcriptional activator proteins with zinc finger (ZFP) motifs. We tested a panel of custom-designed ZFP for their ability to activate the utrophin A promoter. Expression of one such ZFP efficiently increased, in a time-dependent manner, utrophin transcript and protein levels both in vitro and in vivo. In dystrophic mouse (mdx) muscles, administration of adenoviral vectors expressing this ZFP led to significant enhancement of muscle function with decreased necrosis, restoration of the dystrophin-associated proteins, and improved resistance to eccentric contractions. These studies provide evidence that specifically designed ZFPs can act as strong transcriptional activators of the utrophin A promoter. These may thus serve as attractive therapeutic agents for dystrophin deficiency states such as Duchenne muscular dystrophy.


Assuntos
Regulação da Expressão Gênica , Músculos/patologia , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Regulação para Cima , Utrofina/genética , Animais , Humanos , Camundongos , Camundongos Transgênicos , Contração Muscular , Músculos/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Necrose , Fatores de Tempo
19.
Mol Ther ; 15(10): 1767-74, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17667948

RESUMO

Helper-dependent adenovirus vector (AdV)-mediated full-length dystrophin expression leads to significant mitigation of the dystrophic phenotype of the mdx mouse. However, dystrophin, as a neoantigen, elicits antibody formation. As an alternative approach, we evaluated gene transfer of full-length murine utrophin, a functional homologue of dystrophin that is normally present only at the neuromuscular junction. A single injection in the tibialis anterior (TA) muscle of the helper-dependent adenovirus vector encoding utrophin provided very good transduction, with 58% of fibers demonstrating sarcolemmal utrophin expression in the neonates, and 35% utrophin-positive (Utr(+)) fibers in adults. The presence of utrophin prevented extensive necrosis in the neonates, halted further necrosis in the adults, and led to restoration of sarcolemmal expression of dystrophin-associated proteins up to 1 year after injection. Marked physiological improvement was observed in both neonates and adults. Neither increased humoral responses nor cellular immune responses were evident. However, there was a time-related decline of the initial high utrophin expression. Although viral DNA persisted in animals that were injected in the neonatal stage, viral DNA levels decreased in muscles of adult mice. These results demonstrate that although utrophin gene transfer leads to amelioration of the dystrophic phenotype, the effects are not sustained upon loss of utrophin expression.


Assuntos
Adenoviridae/genética , Distrofina/genética , Utrofina/genética , Animais , Animais Recém-Nascidos , Formação de Anticorpos , DNA Viral/metabolismo , Imunidade Celular , Camundongos , Camundongos Mutantes , Músculo Esquelético/metabolismo , Reação em Cadeia da Polimerase , Transdução Genética , Utrofina/administração & dosagem , Utrofina/imunologia
20.
Neurology ; 66(7): 1114-6, 2006 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-16606933

RESUMO

The authors report a genotype-phenotype correlation in a limb-girdle muscular dystrophy 2B family. Two severely affected sisters were homozygous for a dysferlin null mutation. Their mildly affected compound heterozygous mother harbored, in addition to one null allele, an in-frame exon-skipping allele caused by a novel lariat branch point mutation. The dysferlin molecule arising from the latter allele appeared to partially complement the null mutation, likely accounting for the mother's mild phenotype.


Assuntos
Proteínas de Membrana/genética , Proteínas Musculares/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação Puntual , Idoso , Disferlina , Feminino , Humanos , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA