Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Brain Dev ; 31(2): 169-72, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18430534

RESUMO

Duchenne muscular dystrophy (DMD) is caused by mutation of the dystrophin gene. Cases of dystrophinopathy with a 2-bp deletion in the dystrophin gene commonly result in DMD. We report here a case of dystrophinopathy in a 9-years-old boy with a 2-bp deletion in exon 74 of the dystrophin gene; however, the boy had no clear clinical signs of muscle weakness. Immunohistochemical studies with N-terminal (DYS3) and rod-domain anti-dystrophin (DYS1) antibodies revealed that the dystrophin signals were weaker than in the control sample (non-dystrophinopathy) at the sarcolemma of myofibers, and the studies with C-terminus anti-dystrophin antibody (DYS2) were negative. Our patient's mutation is located between the binding sites of alpha-syntrophin and alpha-dystrobrevin. These results suggest that this mutation does not clearly induce muscle weakness at least through the age of 9 years.


Assuntos
Distrofina/genética , Debilidade Muscular/genética , Deleção de Sequência , Sequência de Bases , Sítios de Ligação , Western Blotting , Proteínas de Ligação ao Cálcio/metabolismo , Criança , Distrofina/metabolismo , Proteínas Associadas à Distrofina/metabolismo , Éxons , Genes , Humanos , Imuno-Histoquímica , Masculino , Proteínas de Membrana/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Debilidade Muscular/fisiopatologia , Sarcolema/metabolismo
2.
J Child Neurol ; 22(7): 901-6, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17715288

RESUMO

One cause of X-linked dilated cardiomyopathies is mutation of the dystrophin gene. We report the case of a young boy who suffered from dilated cardiomyopathy caused only by dystrophin-deficient cardiac muscle, but who did not present with any clinical signs of skeletal myopathy. Sequence analysis of the patient's dystrophin gene revealed the presence of a novel single point mutation at the first exon-intron boundary, inactivating the 5' splice site consensus sequence of the first intron. The lack of muscle weakness observed clinically can be explained by expression of the brain and Purkinje dystrophin isoforms in skeletal muscle.


Assuntos
Cardiomiopatia Dilatada/genética , Sequência Consenso/genética , Distrofina/genética , Mutação Puntual/genética , Sítios de Splice de RNA/genética , Adolescente , Distrofina/metabolismo , Humanos , Masculino , Músculo Esquelético/metabolismo , Miocárdio/metabolismo , Miocárdio/patologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
3.
Biochem Biophys Res Commun ; 357(4): 957-63, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17466266

RESUMO

We established genetically engineered ES (ZHTc6-MyoD) cells that harbor a tetracycline-regulated expression vector encoding myogenic transcriptional factor MyoD, for the therapy of muscle diseases, especially Duchenne muscular dystrophy (DMD). Almost all the ZHTc6-MyoD cells were induced into muscle lineage after removal of tetracycline. The undifferentiated ZHTc6-MyoD cells are Sca-1+ and c-kit+, but CD34-, all well-known markers for mouse hematopoietic stem cells. In addition, they are able to maintain themselves in the undifferentiated state, even after one month of culture. Therefore, it is possible to obtain a large quantity of ZHTc6-MyoD cells in the undifferentiated state that maintain the potential to differentiate only into muscle lineage. Additionally, at two weeks post-injection of these cells into muscle of mdx, a model mouse of DMD, clusters of dystrophin-positive myofibers were observed at the injection site. Therefore, ES cells have considerable therapeutic potential for treating muscle diseases.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Engenharia Genética/métodos , Células Musculares/citologia , Células Musculares/metabolismo , Proteína MyoD/genética , Animais , Células Cultivadas , Camundongos , Engenharia Tecidual/métodos
4.
Hum Mol Genet ; 15(14): 2266-75, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16777842

RESUMO

Duchenne muscular dystrophy (DMD) is a fatal X-linked muscle-wasting disease caused by mutations of the gene encoding the cytoskeletal protein dystrophin. Therapeutic options for DMD are limited because the pathogenetic mechanism by which dystrophin deficiency produces the clinical phenotype remains obscure. Recent reports of abnormal alpha-adrenergic vasoregulation in the exercising muscles of DMD patients and in the mdx mouse, an animal model of DMD, prompted us to hypothesize that the dystrophin-deficient smooth muscle contributes to the vascular and dystrophic phenotypes of DMD. To test this, we generated transgenic mdx mice that express dystrophin only in smooth muscle (SMTg/mdx). We found that alpha-adrenergic vasoconstriction was markedly attenuated in the contracting hindlimbs of C57BL/10 wild-type mice, an effect that was mediated by nitric oxide (NO) and was severely impaired in the mdx mice. SMTg/mdx mice showed an intermediate phenotype, with partial restoration of the NO-dependent modulation of alpha-adrenergic vasoconstriction in active muscle. In addition, the elevated serum creatine kinase levels observed in mdx mice were significantly reduced in SMTg/mdx mice. This is the first report of a functional role of dystrophin in vascular smooth muscle.


Assuntos
Distrofina/genética , Distrofina/fisiologia , Músculo Liso Vascular/fisiopatologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/fisiopatologia , Animais , Sequência de Bases , Creatina Quinase/sangue , DNA Complementar/genética , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Contração Muscular , Músculo Liso Vascular/irrigação sanguínea , Distrofia Muscular Animal/patologia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/fisiopatologia , Óxido Nítrico Sintase/antagonistas & inibidores , Receptores Adrenérgicos alfa/fisiologia , Vasoconstrição
5.
Brain Dev ; 28(7): 420-5, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16584859

RESUMO

MyoD, a master regulatory gene for myogenesis, converts mesoderm derived cells to the skeletal muscle phenotype MyoD gene transfer into skin fibroblasts has been attempted in an effort to diagnose genetic muscle diseases. Although the gene transduction efficiency of adenoviral gene delivery systems is higher than that of various other systems, the rate of myo-conversion is insignificant. Since high adenovirus doses are cytotoxic and exogenous MyoD expression is insufficient for skin fibroblasts to re-differentiate into muscle cells, we constructed the novel adeno-MyoD vector, Ad.CAGMyoD using the recombinant CAG promoter. Even at a lower multiplicities of infection most skin fibroblasts infected with Ad.CAGMyoD could convert into myotubes without vector-induced cytotoxicity. The converted cells expressed muscle-specific desmin and full-length dystrophin, both of which were detected by Western blotting. Genetic and immunohistochemical analyses using skin fibroblasts and our vector system are reliable and useful for the clinical diagnosis of genetic muscle diseases.


Assuntos
Adenoviridae/fisiologia , Fibroblastos/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Proteína MyoD/genética , Western Blotting/métodos , Células Cultivadas , Pré-Escolar , Desmina/metabolismo , Fibroblastos/patologia , Técnicas de Transferência de Genes , Vetores Genéticos/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica/métodos , Proteína MyoD/metabolismo , Fatores de Tempo
6.
Brain Dev ; 27(6): 400-5, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16122626

RESUMO

Aminoglycoside antibiotics have been found to suppress nonsense mutations located in the defective dystophin gene in mdx mice, suggesting a possible treatment for Duchenne muscular dystrophy (DMD). However, it is very difficult to find patients that are applicable for this therapy, because: (1) only 5-13% of DMD patients have nonsense mutations in the dystrophin gene, (2) it is challenging to find nonsense mutations in the gene because dystrophin cDNA is very long (14 kb), and (3) the efficiency of aminoglycoside-induced read-through is dependent on the kind of nonsense mutation. In order to develop a system for identifying candidates that qualify for aminoglycoside therapy, fibroblasts from nine DMD patients with nonsense mutation of dystrophin gene were isolated, induced to differentiate to myogenic lineage by AdMyoD, and exposed with gentamicin. The dystrophin expression in gentamicin-exposed myotubes was monitored by in vitro dystrophin staining and western blotting analysis. The results showed that gentamicin was able to induce dystrophin expression in the differentiated myotubes by the read-through of the nonsense mutation TGA in the gene; a read-through of the nonsense mutations TAA and TAG did not occur and consequently did not lead to dystrophin expression. Therefore, it is speculated that the aminoglycoside treatment is far more effective for DMD patients that have nonsense mutation TGA than for patients that have nonsense mutation TAA and TAG. In this study, we introduce an easy system to identify patients for this therapy and report for the first time, that dystrophin expression was detected in myotubes of DMD patients using gentamicin.


Assuntos
Antibacterianos/farmacologia , Distrofina/genética , Testes Genéticos/métodos , Gentamicinas/farmacologia , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Western Blotting , Encéfalo , Células Cultivadas , Códon sem Sentido , Códon de Terminação , Distrofina/química , Distrofina/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Expressão Gênica/efeitos dos fármacos , Humanos , Isomerismo , Músculo Esquelético , Células de Purkinje
7.
Epilepsia ; 46 Suppl 5: 152-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15987271

RESUMO

PURPOSE: To evaluate antibody-mediated and cytotoxic T cell-mediated pathogenicity that has been implicated as the autoimmune pathophysiological mechanism in Rasmussen's encephalitis. METHODS: We examined autoantibodies against the N-methyl-d-aspartate glutamate receptor (NMDA-type GluR) epsilon2 subunit and its epitopes in serum and CSF samples from 20 patients [five histologically proven (definitive) Rasmussen's encephalitis with epilepsia partialis continua (EPC), four definitive Rasmussen's encephalitis without EPC, and 11 clinical Rasmussen's encephalitis with EPC]. We examined 3H-thymidine uptake into lymphocytes after stimulation by GluRs. RESULTS: All nine definitive patients (five patients with EPC and four without EPC), and 10 of 11 clinical Rasmussen's encephalitis patients had the autoantibodies. In four patients, the autoantibodies were absent in early stage when epileptic seizures had already become frequent, and appeared subsequently. In two patients, the autoantibodies persisted in the serum after frontal lobe resection or functional hemispherectomy, although epileptic seizures were completely controlled. Autoantibodies to the C2 epitope predominated, while autoantibodies to the extracellular N epitope were rare. The mean 3H-thymidine uptake ratios (stimulation by GluRepsilon2-containing homogenates/stimulation by PHA) were significantly higher in definitive and clinical Rasmussen encephalitis patients than in controls. The mean 3H-thymidine uptake ratios (relative to PHA) were significantly higher for GluRepsilon2-containing homogenate than for control homogenate or GluRdelta2-containing homogenate. CONCLUSIONS: Autoantibodies against GluRepsilon2 may be one of the diagnostic markers for Rasmussen's encephalitis with and without EPC. Patients have activated T cells stimulated by GluRepsilon2 in peripheral blood circulation. We speculate that cellular autoimmunity and the subsequent humoral autoimmunity against GluRepsilon2 may contribute to the pathophysiological processes in Rasmussen's encephalitis.


Assuntos
Autoanticorpos/imunologia , Encefalite/imunologia , Epilepsia Parcial Contínua/imunologia , Imunidade Celular/imunologia , Receptores de N-Metil-D-Aspartato/imunologia , Adolescente , Adulto , Autoanticorpos/sangue , Autoanticorpos/líquido cefalorraquidiano , Autoimunidade/imunologia , Biomarcadores , Criança , Encefalite/sangue , Encefalite/líquido cefalorraquidiano , Encefalite/diagnóstico , Epilepsia Parcial Contínua/sangue , Epilepsia Parcial Contínua/líquido cefalorraquidiano , Feminino , Humanos , Masculino , Receptores de N-Metil-D-Aspartato/análise , Receptores de N-Metil-D-Aspartato/sangue
8.
J Gene Med ; 6(2): 155-65, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14978769

RESUMO

BACKGROUND: Adenoviral gene transfer to adult skeletal muscle is hindered by several major limitations, including host immune responses and maturation-dependent loss of myofiber infectivity. Ex vivo gene delivery is more efficient than direct viral injection in surmounting maturation-dependent adenoviral transduction. Here we investigated the use of helper cells to improve the efficiency of ex vivo gene transfer to adult mouse skeletal muscle. METHODS: New producer cells carrying the E1 gene of adenovirus type 5 (E32 cells) were developed using primary myoblasts from mdx mice. The E32 cells and 293 cells were infected with an E1-deleted first-generation adenovirus carrying the LacZ gene. These transduced helper cells were injected into the skeletal muscle of adult mdx and SCID mice. RESULTS: LacZ-positive mature myofibers were detected in the skeletal muscle of adult mice sacrificed 5 days post-injection. The gene transfer efficiency using 293 cells and E32 cells was 6.2 and 3.6 times higher than myoblast-mediated gene transfer, respectively. Ex vivo gene transfer of these cell types led to a better outcome than did direct adenoviral injection. CONCLUSIONS: We achieved more efficient adenoviral gene transduction by using 293 and E32 helper cells than by myoblast-mediated gene transfer and direct viral injection. These helper cells also enabled adenoviral gene transfer to mature myofibers. The mechanisms by which this method facilitated adenoviral gene transfer to mature myofibers remains unclear; however, we hypothesize that the in vivo occurrence of cytopathic effects (CPE) in the transduced 293 and E32 helper cell populations facilitated the improved adenoviral transduction of myofibers.


Assuntos
Adenoviridae , Terapia Genética , Vetores Genéticos , Vírus Auxiliares , Músculo Esquelético/metabolismo , Animais , Fusão Celular , Genes Reporter , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos mdx , Distrofia Muscular de Duchenne/terapia
9.
Hum Gene Ther ; 14(16): 1535-46, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-14577915

RESUMO

Duchenne's muscular dystrophy (DMD) is a lethal muscle disease caused by a lack of dystrophin expression at the sarcolemma of muscle fibers. We investigated retroviral vector delivery of dystrophin in dystrophin-deficient DMD(mdx) (hereafter referred to as mdx) mice via an ex vivo approach using mdx muscle-derived stem cells (MDSCs). We generated a retrovirus carrying a functional human mini-dystrophin (RetroDys3999) and used it to stably transduce mdx MDSCs obtained by the preplate technique (MD3999). These MD3999 cells expressed dystrophin and continued to express stem cell markers, including CD34 and Sca-1. MD3999 cells injected into mdx mouse skeletal muscle were able to deliver dystrophin. Though a relatively low number of dystrophin-positive myofibers was generated within the gastrocnemius muscle, these fibers persisted for up to 24 weeks postinjection. The injection of cells from additional MDSC/Dys3999 clones into mdx skeletal muscle resulted in varying numbers of dystrophin-positive myofibers, suggesting a differential regenerating capacity among the clones. At 2 and 4 weeks postinjection, the infiltration of CD4- and CD8-positive lymphocytes and a variety of cytokines was detected within the injected site. These data suggest that the transplantation of retrovirally transduced mdx MDSCs can enable persistent dystrophin restoration in mdx skeletal muscle; however, the differential regenerating capacity observed among the MDSC/Dys3999 clones and the postinjection immune response are potential challenges facing this technology.


Assuntos
Distrofina/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Transplante de Células-Tronco , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Transplante de Células , Células Cultivadas , Distrofina/deficiência , Expressão Gênica , Terapia Genética , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Músculo Esquelético/imunologia , Retroviridae/genética , Transplante Isogênico
10.
Nat Cell Biol ; 5(7): 640-6, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12792651

RESUMO

Muscle-derived stem cells (MDSCs) can differentiate into multiple lineages, including haematopoietic lineages. However, it is unknown whether MDSCs preserve their myogenic potential after differentiation into other lineages. To address this issue, we isolated from dystrophic muscle a population of MDSCs that express stem-cell markers and can differentiate into various lineages. After systemic delivery of three MDSC clones into lethally irradiated mice, we found that differentiation of the donor cells into various lineages of the haematopoietic system resulted in repopulation of the recipients' bone marrow. Donor-derived bone-marrow cells, isolated from these recipients by fluorescence-activated cell sorting (FACS), also repopulated the bone marrow of secondary, lethally irradiated, recipients and differentiated into myogenic cells both in vitro and in vivo in normal mdx mice. These findings demonstrate that MDSC clones retain their myogenic potential after haematopoietic differentiation.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células Clonais/citologia , Células-Tronco Hematopoéticas/citologia , Músculo Esquelético/embriologia , Músculo Esquelético/crescimento & desenvolvimento , Células-Tronco/citologia , Animais , Biomarcadores , Células da Medula Óssea/efeitos da radiação , Células Cultivadas , Células Clonais/transplante , Citometria de Fluxo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Mioblastos/citologia , Mioblastos/fisiologia , Células-Tronco/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...