Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Hum Mol Genet ; 23(7): 1842-55, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24234655

RESUMO

Mutations in fukutin-related protein (FKRP) underlie a group of muscular dystrophies associated with the hypoglycosylation of α-dystroglycan (α-DG), a proportion of which show central nervous system involvement. Our original FKRP knock-down mouse (FKRP(KD)) replicated many of the characteristics seen in patients at the severe end of the dystroglycanopathy spectrum but died perinatally precluding its full phenotyping and use in testing potential therapies. We have now overcome this by crossing FKRP(KD) mice with those expressing Cre recombinase under the Sox1 promoter. Owing to our original targeting strategy, this has resulted in the restoration of Fkrp levels in the central nervous system but not the muscle, thereby generating a new model (FKRP(MD)) which develops a progressive muscular dystrophy resembling what is observed in limb girdle muscular dystrophy. Like-acetylglucosaminyltransferase (LARGE) is a bifunctional glycosyltransferase previously shown to hyperglycosylate α-DG. To investigate the therapeutic potential of LARGE up-regulation, we have now crossed the FKRP(MD) line with one overexpressing LARGE and show that, contrary to expectation, this results in a worsening of the muscle pathology implying that any future strategies based upon LARGE up-regulation require careful management.


Assuntos
Distroglicanas/metabolismo , N-Acetilglucosaminiltransferases/biossíntese , N-Acetilglucosaminiltransferases/genética , Proteínas/genética , Síndrome de Walker-Warburg/genética , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Glicosilação , Laminina/biossíntese , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Mutação , Pentosiltransferases , Transferases , Regulação para Cima , Síndrome de Walker-Warburg/mortalidade
2.
Stem Cells ; 30(10): 2330-41, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22887880

RESUMO

The dystrophin-associated glycoprotein complex (DGC) is found at the muscle fiber sarcolemma and forms an essential structural link between the basal lamina and internal cytoskeleton. In a set of muscular dystrophies known as the dystroglycanopathies, hypoglycosylation of the DGC component α-dystroglycan results in reduced binding to basal lamina components, a loss in structural stability, and repeated cycles of muscle fiber degeneration and regeneration. The satellite cells are the key stem cells responsible for muscle repair and reside between the basal lamina and sarcolemma. In this study, we aimed to determine whether pathological changes associated with the dystroglycanopathies affect satellite cell function. In the Large(myd) mouse dystroglycanopathy model, satellite cells are present in significantly greater numbers but display reduced proliferation on their native muscle fibers in vitro, compared with wild type. However, when removed from their fiber, proliferation in culture is restored to that of wild type. Immunohistochemical analysis of Large(myd) muscle reveals alterations to the basal lamina and interstitium, including marked disorganization of laminin, upregulation of fibronectin and collagens. Proliferation and differentiation of wild-type satellite cells is impaired when cultured on substrates such as collagen and fibronectin, compared with laminins. When engrafted into irradiated tibialis anterior muscles of mdx-nude mice, wild-type satellite cells expanded on laminin contribute significantly more to muscle regeneration than those expanded on fibronectin. These results suggest that defects in α-dystroglycan glycosylation are associated with an alteration in the satellite cell niche, and that regenerative potential in the dystroglycanopathies may be perturbed.


Assuntos
Membrana Basal/metabolismo , Distroglicanas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Animal/metabolismo , Sarcolema/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Membrana Basal/patologia , Diferenciação Celular , Proliferação de Células , Colágeno/química , Colágeno/metabolismo , Modelos Animais de Doenças , Fibronectinas/química , Fibronectinas/metabolismo , Glicosilação , Humanos , Laminina/química , Laminina/metabolismo , Camundongos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Distrofia Muscular Animal/patologia , Ligação Proteica , Sarcolema/patologia , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/transplante
3.
J Neurosci ; 31(36): 12927-35, 2011 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-21900571

RESUMO

Mutations in fukutin-related protein (FKRP) are responsible for a common group of muscular dystrophies ranging from adult onset limb girdle muscular dystrophies to severe congenital forms with associated structural brain involvement. The defining feature of this group of disorders is the hypoglycosylation of α-dystroglycan and its inability to effectively bind extracellular matrix ligands such as laminin α2. However, α-dystroglycan has the potential to interact with a number of laminin isoforms many of which are basement membrane/tissue specific and developmentally regulated. To further investigate this we evaluated laminin α-chain expression in the cerebral cortex and eye of our FKRP knock-down mouse (FKRP(KD)). These mice showed a marked disturbance in the deposition of laminin α-chains including α1, α2, α4, and α5, although only laminin α1- and γ1-chain mRNA expression was significantly upregulated relative to controls. Moreover, there was a diffuse pattern of laminin deposition below the pial surface which correlated with an abrupt termination of many of the radial glial cells. This along with the pial basement membrane defects, contributed to the abnormal positioning of both early- and late-born neurons. Defects in the inner limiting membrane of the eye were associated with a reduction of laminin α1 demonstrating the involvement of the α-dystroglycan:laminin α1 axis in the disease process. These observations demonstrate for the first time that a reduction in Fkrp influences the ability of tissue-specific forms of α-dystroglycan to direct the deposition of several laminin isoforms in the formation of different basement membranes.


Assuntos
Química Encefálica/genética , Olho/metabolismo , Laminina/metabolismo , Proteínas/fisiologia , Animais , Apoptose/fisiologia , Membrana Basal/efeitos dos fármacos , Membrana Basal/metabolismo , Movimento Celular/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Distroglicanas/metabolismo , Glicosilação , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Neurônios/fisiologia , Pentosiltransferases , Fenótipo , Proteínas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/fisiologia , Transferases
4.
Curr Opin Neurol ; 24(5): 437-42, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21825985

RESUMO

PURPOSE OF REVIEW: Dystroglycanopathies are a common group of diseases characterized by a reduction in α-dystroglycan glycosylation. This review discusses the recent novel discovery of additional dystroglycanopathy variants and progress in dystroglycanopathy animal models. RECENT FINDINGS: Several novel glycosyltransferase genes have been found to be responsible for a dystroglycanopathy phenotype, and in addition recessive mutations in DAG1 have been identified for the first time in a primary dystroglycanopathy. Studies in dystroglycanopathy mouse models have clarified some aspects of the structural defects observed in the central nervous system and in the eye, whereas a study in zebrafish implicates unfolded protein response in the pathogenesis of two of the secondary dystroglycanopathies. SUMMARY: Improved understanding of the molecular bases of dystroglycanopathies will lead to more precise diagnosis and genetic counseling; therapeutic strategies are being developed and tested in the preclinical models and it is hoped that these observations will pave the way to therapeutic interventions in humans.


Assuntos
Distroglicanas/metabolismo , Distrofias Musculares/metabolismo , Síndrome de Walker-Warburg/metabolismo , Animais , Modelos Animais de Doenças , Distroglicanas/genética , Glicosilação , Humanos , Camundongos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/terapia , Síndrome de Walker-Warburg/genética , Síndrome de Walker-Warburg/terapia , Peixe-Zebra
5.
J Neuropathol Exp Neurol ; 79(12): 1257-1264, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-33051673

RESUMO

Mutations in the fukutin-related protein gene, FKRP, are the most frequent single cause of α-dystroglycanopathy. Rare FKRP mutations are clinically not well characterized. Here, we review the phenotype associated with the rare c.919T>A mutation in FKRP in humans and mice. We describe clinical and paraclinical findings in 6 patients, 2 homozygous, and 4-compound heterozygous for c.919T>A, and compare findings with a mouse model we generated, which is homozygous for the same mutation. In patients, the mutation at the homozygous state is associated with a severe congenital muscular dystrophy phenotype invariably characterized by severe multisystem disease and early death. Compound heterozygous patients have a severe limb-girdle muscular dystrophy phenotype, loss of ambulation before age 20 and respiratory insufficiency. In contrast, mice homozygous for the same mutation show no symptoms or signs of muscle disease. Evidence therefore defines the FKRP c.919T>A as a very severe mutation in humans. The huge discrepancy between phenotypes in humans and mice suggests that differences in protein folding/processing exist between human and mouse Fkrp. This emphasizes the need for more detailed structural analyses of FKRP and shows the challenges of developing appropriate animal models of dystroglycanopathies that mimic the disease course in humans.


Assuntos
Mutação , Pentosiltransferases/genética , Fenótipo , Síndrome de Walker-Warburg/genética , Animais , Modelos Animais de Doenças , Humanos , Camundongos
6.
Arch Neurol ; 65(1): 137-41, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18195152

RESUMO

BACKGROUND: Mutations in protein-O-mannose-beta1,2-N-acetylglucosaminyltransferase 1 (POMGnT1) have been found in muscle-eye-brain disease, a congenital muscular dystrophy with structural eye and brain defects and severe mental retardation. OBJECTIVE: To investigate whether mutations in POMGnT1 could be responsible for milder allelic variants of muscular dystrophy. DESIGN: Screening for mutations in POMGnT1. SETTING: Tertiary neuromuscular unit. PATIENT: A patient with limb-girdle muscular dystrophy phenotype, with onset at 12 years of age, severe myopia, normal intellect, and decreased alpha-dystroglycan immunolabeling in skeletal muscle. RESULTS: A homozygous POMGnT1 missense mutation (c.1666G>A, p.Asp556Asn) was identified. Enzyme studies of the patient's fibroblasts showed an altered kinetic profile, less marked than in patients with muscle-eye-brain disease and in keeping with the relatively mild phenotype in our patient. CONCLUSIONS: Our findings widen the spectrum of disorders known to result from mutations in POMGnT1 to include limb-girdle muscular dystrophy with no mental retardation. We propose that this condition be known as LGMD2M. The enzyme assay used to diagnose muscle-eye-brain disease may not detect subtle abnormalities of POMGnT1 function, and additional kinetic studies must be carried out in such cases.


Assuntos
Distrofia Muscular do Cíngulo dos Membros/genética , N-Acetilglucosaminiltransferases/genética , Alelos , Western Blotting , Criança , Análise Mutacional de DNA , Distroglicanas/metabolismo , Fibroblastos/enzimologia , Testes Genéticos , Humanos , Imuno-Histoquímica , Cinética , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/complicações , Distrofia Muscular do Cíngulo dos Membros/psicologia , Mutação , Mutação de Sentido Incorreto/genética , Miopia/etiologia , Fenótipo
7.
Biochem Soc Trans ; 36(Pt 6): 1335-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19021551

RESUMO

EDMD (Emery-Dreifuss muscular dystrophy) is caused by mutations in either the gene encoding for lamin A/C (LMNA) located at 1q21.2-q21.3 or emerin (EMD) located at Xq28. Autosomal dominant EDMD caused by LMNA mutations is more common than the X-linked form and often more severe, with an earlier onset. At the histological and histochemical levels, both X-linked and autosomal dominant EDMD appear similar. However, individuals with the same genetic disorder often show remarkable differences in clinical severity, a finding generally attributed to the genetic background. The clinical and pathological findings in EDMD patients found to have mutations in more than one gene are also discussed. There is now much interest in the phenotype of several animal models for EDMD which should lead to an increased insight into the pathogenesis of this disorder, particularly that relating to the heart phenotype.


Assuntos
Distrofia Muscular de Emery-Dreifuss/patologia , Humanos , Distrofia Muscular de Emery-Dreifuss/genética , Mutação/genética
8.
Brain ; 130(Pt 10): 2725-35, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17878207

RESUMO

Muscular dystrophies with reduced glycosylation of alpha-dystroglycan (alpha-DG), commonly referred to as dystroglycanopathies, are a heterogeneous group of autosomal recessive conditions which include a wide spectrum of clinical severity. Reported phenotypes range from severe congenital onset Walker-Warburg syndrome (WWS) with severe structural brain and eye involvement, to relatively mild adult onset limb girdle muscular dystrophy (LGMD). Specific clinical syndromes were originally described in association with mutations in any one of six demonstrated or putative glycosyltransferases. Work performed on patients with mutations in the FKRP gene has identified that the spectrum of phenotypes due to mutations in this gene is much wider than originally assumed. To further define the mutation frequency and phenotypes associated with mutations in the other five genes, we studied a large cohort of patients with evidence of a dystroglycanopathy. Exclusion of mutations in FKRP was a prerequisite for participation in this study. Ninety-two probands were screened for mutations in POMT1, POMT2, POMGnT1, fukutin and LARGE. Homozygous and compound heterozygous mutations were detected in a total of 31 probands (34 individuals from 31 families); 37 different mutations were identified, of which 32 were novel. Mutations in POMT2 were the most prevalent in our cohort with nine cases, followed by POMT1 with eight cases, POMGnT1 with seven cases, fukutin with six cases and LARGE with only a single case. All patients with POMT1 and POMT2 mutations had evidence of either structural or functional central nervous system involvement including four patients with mental retardation and a LGMD phenotype. In contrast mutations in fukutin and POMGnT1 were detected in four patients with LGMD and no evidence of brain involvement. The majority of patients (six out of nine) with mutations in POMT2 had a Muscle-Eye-Brain (MEB)-like condition. In addition we identified a mutation in the gene LARGE in a patient with WWS. Our data expands the clinical phenotypes associated with POMT1, POMT2, POMGnT1, fukutin and LARGE mutations. Mutations in these five glycosyltransferase genes were detected in 34% of patients indicating that, after the exclusion of FKRP, the majority of patients with a dystroglycanopathy harbour mutations in novel genes.


Assuntos
Distroglicanas/genética , Distrofias Musculares/genética , Mutação , Pré-Escolar , Estudos de Coortes , Análise Mutacional de DNA , Distroglicanas/metabolismo , Glicosilação , Humanos , Lactente , Masculino , Manosiltransferases/genética , Proteínas de Membrana/genética , Distrofias Musculares/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , N-Acetilglucosaminiltransferases/genética , Fenótipo
9.
Am J Vet Res ; 69(12): 1637-45, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19046012

RESUMO

OBJECTIVE: To develop a reliable method for converting cultured equine skin-derived fibroblasts into muscle cells. SAMPLE POPULATION: Equine skin-derived fibroblasts. PROCEDURES: The equine myogenic differentiation 1 (eqMyoD) genomic sequence was obtained by use of equine bacterial artificial chromosome screening and PCR sequencing. Total mRNA was extracted from foal skeletal muscle, and eqMyoD cDNA was cloned into a plasmid vector with an internal ribosomal entry site to express bicistronic eqMyoD or enhanced green fluorescent protein (EGFP). Transient expression was confirmed by immunocytochemical analysis and western immunoblots in equine fibroblasts and fibroblasts from National Institutes of Health Swiss mouse embryos, prior to generation of a lentiviral vector containing the same coding sequences. Transformation of equine skin-derived cells into skeletal myotubes was examined by use of immunohistochemical analysis, western immunoblotting, and periodic acid-Schiff staining. RESULTS: eqMyoD mRNA consists of 960 bp and shares high homology with myogenic differentiation 1 from other mammals. Transfection confirmed the expression of a 53-kd protein with mainly nuclear localization. Lentiviral transduction was efficient, with approximately 80% of EGFP-positive cells transformed into multinucleated myotubes during 15 days, as determined by expression of the muscle-specific proteins desmin, troponin-T, and sarcomeric myosin and by cytoplasmic storage of glycogen. CONCLUSIONS AND CLINICAL RELEVANCE: Equine primary fibroblasts were transformed by lentiviral transduction of eqMyoD into fusion-competent myoblasts. This may offer a preferable alternative to primary myoblast cultures for the investigation of cellular defects associated with muscle diseases of horses, such as recurrent exertional rhabdomyolysis and polysaccharide storage myopathy.


Assuntos
Fibroblastos/citologia , Cavalos , Lentivirus/fisiologia , Fibras Musculares Esqueléticas/citologia , Proteína MyoD/metabolismo , Pele/citologia , Células 3T3 , Sequência de Aminoácidos , Animais , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/virologia , Proteína MyoD/genética
10.
Neuromuscul Disord ; 17(4): 297-305, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17329105

RESUMO

Autosomal dominant Emery-Dreifuss muscular dystrophy (AD-EDMD) is one of a number of allelic disorders caused by mutations in the nuclear lamina proteins, lamins A and C. The disorder is characterised by the early onset of skeletal muscle weakness and joint contractures and later, by dilated cardiomyopathy and cardiac arrythmias. Although the pathophysiology is not understood, one theory suggests that disordered structural organisation at weakened nuclei in contractile cells may underlie the disease. Previous work shows that mice deficient in lamin A/C develop similar skeletal and cardiac muscle signs to patients with AD-EDMD and ultrastructural examination of muscle from these mice shows abnormal localisation of desmin. We hypothesised therefore that desmin localisation may be abnormal in muscle or cells from patients with AD-EDMD and/or in cells expressing mutant lamins. In order to evaluate this, desmin immunolocalisation was determined in skeletal muscle biopsy sections from patients with AD-EDMD and cell lines including MyoD-transfected fibroblast-derived myotubes from AD-EDMD patients and murine embryonic stem cell-derived cardiomyocytes stably transfected with mutant human lamin A. Ultrastructural examination of patient muscle was also performed. Desmin was expressed and localised normally in patient muscle and cell lines and ultrastructural examination was similar to controls. These results fail to provide any evidence that dominant mutations in lamin A/C lead to a disorganisation of the desmin associated cytoskeleton.


Assuntos
Desmina/metabolismo , Imuno-Histoquímica , Distrofia Muscular de Emery-Dreifuss/metabolismo , Adolescente , Adulto , Diferenciação Celular , Células Cultivadas , Criança , Pré-Escolar , Desmina/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/ultraestrutura , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Microscopia Eletrônica de Transmissão , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/ultraestrutura , Distrofia Muscular de Emery-Dreifuss/patologia , Mutação/fisiologia , Proteína MyoD/farmacologia , Transfecção/métodos
12.
Brain Pathol ; 26(4): 465-78, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26306834

RESUMO

The secondary dystroglycanopathies are characterized by the hypoglycosylation of alpha dystroglycan, and are associated with mutations in at least 18 genes that act on the glycosylation of this cell surface receptor rather than the Dag1 gene itself. At the severe end of the disease spectrum, there are substantial structural brain defects, the most striking of which is often cobblestone lissencephaly. The aim of this study was to determine the gene-specific aspects of the dystroglycanopathy brain phenotype through a detailed investigation of the structural brain defects present at birth in three mouse models of dystroglycanopathy-the FKRP(KD) , which has an 80% reduction in Fkrp transcript levels; the Pomgnt1null , which carries a deletion of exons 7-16 of the Pomgnt1 gene; and the Large(myd) mouse, which carries a deletion of exons 5-7 of the Large gene. We show a rostrocaudal and mediolateral gradient in the severity of brain lesions in FKRP(KD) , and to a lesser extent Pomgnt1null mice. Furthermore, the mislocalization of Cajal-Retzius cells is correlated with the gradient of these lesions and the severity of the brain phenotype in these models. Overall these observations implicate gene-specific differences in the pathogenesis of brain lesions in this group of disorders.


Assuntos
Encéfalo/patologia , Doenças Neuromusculares/patologia , Animais , Modelos Animais de Doenças , Genótipo , Camundongos , N-Acetilglucosaminiltransferases/genética , Doenças Neuromusculares/genética , Neurônios/patologia , Pentosiltransferases , Fenótipo , Proteínas/genética , Proteína Reelina , Transferases
13.
Eur J Cell Biol ; 84(9): 765-81, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16218190

RESUMO

Mutations in the LMNA gene, which encodes nuclear lamins A and C by alternative splicing, can give rise to Emery-Dreifuss muscular dystrophy. The mechanism by which lamins A and C separately contribute to this molecular phenotype is unknown. To address this question we examined ten LMNA mutations exogenously expressed as lamins A and C in COS-7 cells. Eight of the mutations when expressed in lamin A, exhibited a range of nuclear mislocalisation patterns. However, two mutations (T150P and delQ355) almost completely relocated exogenous lamin A from the nuclear envelope to the cytoplasm, disrupted nuclear envelope reassembly following cell division and altered the protein composition of the mid-body. In contrast, exogenously expressed DsRed2-tagged mutant lamin C constructs were only inserted into the nuclear lamina if co-expressed with any EGFP-tagged lamin A construct, except with one carrying the T150P mutation. The T150P, R527P and L530P mutations reduced the ability of lamin A, but not lamin C from binding to emerin. These data identify specific functional roles for the emerin-lamin C- and emerin-lamin A- containing protein complexes and is the first report to suggest that the A-type lamin mutations may be differentially dysfunctional for the same LMNA mutation.


Assuntos
Lamina Tipo A/genética , Distrofia Muscular de Emery-Dreifuss/metabolismo , Animais , Células COS , DNA Complementar/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia de Fluorescência , Mutação , Lâmina Nuclear/metabolismo , Proteínas Nucleares , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Timopoietinas/genética , Timopoietinas/metabolismo , Transfecção
14.
Neuromuscul Disord ; 15(12): 836-43, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16288869

RESUMO

MDC1C and LGMD2I are two allelic forms of muscular dystrophies caused by mutations in the gene encoding for fukutin related protein (FKRP). FKRP encodes for a putative glycosyltransferase, the precise function of which is unknown. However, the marked reduction of alpha-dystroglycan glycosylation in the muscle of MDC1C and LGMD2I patients suggests a role for FKRP in dystroglycan processing. Using a polyclonal antibody raised against FKRP we now show that endogenous FKRP locates to the Golgi apparatus of neuronal, oligodendroglial, and the cardiac muscle cell line H9c2. In differentiated C2C12 myotubes and in transverse sections of normal skeletal and cardiac muscle, endogenous FKRP surrounded the myonuclei. This localisation was unaffected in the skeletal muscle of patients with MDC1C and LGMD2I carrying various FKRP mutations. These observations imply a specific role for FKRP during striated muscle, neuronal and glial development and suggest that protein mis-localisation is not a common mechanism of disease in FKRP-related dystrophies.


Assuntos
Músculo Esquelético/patologia , Distrofias Musculares , Mutação , Neurônios/patologia , Proteínas/metabolismo , Animais , Autoantígenos , Western Blotting/métodos , Linhagem Celular , Desmina/metabolismo , Feto , Complexo de Golgi/metabolismo , Humanos , Imuno-Histoquímica/métodos , Proteínas de Membrana/metabolismo , Camundongos , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Mioblastos/metabolismo , Mioblastos/patologia , Neuroblastoma , Neurônios/metabolismo , Pentosiltransferases , Ratos , Frações Subcelulares/metabolismo
15.
Neuromuscul Disord ; 25(1): 32-42, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25387694

RESUMO

We generated a novel monoclonal antibody, DAG-6F4, against alpha-dystroglycan which immunolabels the sarcolemma in human muscle biopsies. Its seven amino-acid epitope, PNQRPEL, was identified using phage-displayed peptides and is located immediately after the highly-glycosylated mucin domain of alpha-dystroglycan. On Western blots of recombinant alpha-dystroglycan, epitope accessibility was reduced, but not entirely prevented, by glycosylation. DAG-6F4 immunolabelling was markedly reduced in muscle biopsies from Duchenne muscular dystrophy patients consistent with disruption of the dystroglycan complex. In a range of dystroglycanopathy patients with reduced/altered glycosylation, staining by DAG-6F4 was often less reduced than staining by IIH6 (antibody against the glycan epitope added by LARGE and commonly used to identify glycosylated alpha-dystroglycan). Whereas IIH6 was reduced in all patients, DAG-6F4 was hardly changed in a LARGE patient, less reduced than IIH6 in limb-girdle muscular dystrophy type 2I, but as reduced as IIH6 in some congenital muscular dystrophy patients. Although absence of the LARGE-dependent laminin-binding site appears not to affect alpha-dystroglycan stability at the sarcolemma, the results suggest that further reduction in aDG glycosylation may reduce its stability. These studies suggest that DAG-6F4 may be a useful addition to the antibody repertoire for evaluating the dystroglycan complex in neuromuscular disorders.


Assuntos
Anticorpos Monoclonais/imunologia , Distroglicanas/análise , Distrofia Muscular de Duchenne/patologia , Adulto , Sequência de Aminoácidos , Animais , Pré-Escolar , Distroglicanas/metabolismo , Glicosilação , Células HEK293 , Humanos , Imuno-Histoquímica , Lactente , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Músculo Esquelético/imunologia , Distrofia Muscular de Duchenne/diagnóstico , Sarcolema/imunologia
16.
Neuromuscul Disord ; 13(1): 21-31, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12467729

RESUMO

A lack of effective treatments for Duchenne muscular dystrophy, a fatal X-linked myopathy, has focused attention on the possibility of gene therapy. The aim of the gene therapy approach is the restoration of the dystrophin associated complex of proteins, one member of which is neuronal nitric oxide synthase, an important enzyme in signal transduction. Transgenic mdx mice and plasmid gene transfer of both human and murine recombinant dystrophins was used to assess whether nNOS could be restored to the sarcolemma following dystrophin gene transfer at a variety of levels of expression. Murine revertant fibres and human patients with different dystrophin deletions were used to assess the relationship between exon deletion and loss of neuronal nitric oxide synthase localization to the sarcolemma. We demonstrate that the domain encoded by exons 45-48 is required for localization of neuronal nitric oxide synthase to the sarcolemma. On the basis of these observations we suggest that neuronal nitric oxide synthase is a useful marker for complete restoration of the dystrophin associated complex and should be used as one of the criteria for selecting the recombinant molecule to be used for gene therapy in Duchenne muscular dystrophy.


Assuntos
Distrofina/metabolismo , Músculo Esquelético/enzimologia , Distrofia Muscular Animal/enzimologia , Óxido Nítrico Sintase/metabolismo , Adulto , Animais , Biomarcadores/análise , Proteínas de Ligação ao Cálcio , Proteínas do Citoesqueleto/metabolismo , Éxons , Técnicas de Transferência de Genes , Humanos , Imuno-Histoquímica/métodos , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular de Duchenne/enzimologia , Distrofia Muscular de Duchenne/metabolismo , Distrofia Muscular de Duchenne/patologia , Óxido Nítrico Sintase Tipo I , Fragmentos de Peptídeos/metabolismo , Sarcolema/enzimologia , Utrofina
17.
Eur J Paediatr Neurol ; 7(3): 129-37, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12788039

RESUMO

Walker-Warburg syndrome (WWS) is an autosomal recessive disorder characterized by the combined involvement of the central nervous and skeletal muscle systems. Although the molecular basis of WWS remains unknown, defects in the muscle fibre basal lamina are characteristic of other forms of congenital muscular dystrophy (CMD). In agreement with this, some forms of CMD, due to glycosyltransferase defects, display a reduction in the immunolabelling of alpha-dystroglycan, whilst beta-dystroglycan labelling appears normal. Here we describe an almost complete absence of alpha-dystroglycan using both immunohistochemistry and immunoblotting in two patients with WWS. In addition, there was a mild reduction of laminin-alpha 2. In contrast, immunohistochemical labelling of perlecan and collagen VI was normal. Linkage analysis excluded the recently identified POMT1 locus, responsible for a proportion of WWS cases. These results confirm that WWS is a genetically heterogeneous condition and suggest that disruption of the alpha-dystroglycan/laminin-alpha 2 axis in the basal lamina may play a role in the degeneration of muscle fibres in WWS-also in cases not due to POMT1 defects.


Assuntos
Doenças do Sistema Nervoso Central/complicações , Doenças do Sistema Nervoso Central/metabolismo , Proteínas do Citoesqueleto/metabolismo , Manosiltransferases/metabolismo , Glicoproteínas de Membrana/metabolismo , Distrofias Musculares/complicações , Distrofias Musculares/metabolismo , Membrana Basal/metabolismo , Membrana Basal/patologia , Western Blotting , Doenças do Sistema Nervoso Central/genética , Pré-Escolar , Colágeno Tipo VI/metabolismo , Distroglicanas , Feminino , Ligação Genética/genética , Glicosilação , Proteoglicanas de Heparan Sulfato/metabolismo , Humanos , Imuno-Histoquímica , Lactente , Laminina/deficiência , Masculino , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Distrofias Musculares/genética , Proteínas Quinases/genética , Proteínas de Schizosaccharomyces pombe , Síndrome
18.
Eur J Paediatr Neurol ; 6(6): 309-14, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12401455

RESUMO

We report clinical and muscle magnetic resonance imaging (MRI) findings in three individuals (aged 6, 26 and 73 years) from a three-generation family with Bethlem myopathy, confirmed by molecular genetic analysis which showed an exon skipping mutation in the COL6A1 gene. The clinical severity ranged from mild proximal weakness and distal laxity in the younger patients, to inability to stand or walk and severe contractures in the 76-year-old grandmother. The pattern of muscle involvement showed variable severity in parallel with the severity of motor function impairment. Although there was a marked variability in the severity of the MRI findings, it was possible to recognize a specific pattern of muscle involvement in all three patients. This consisted of involvement of the peripheral region of the vastus lateralis and hamstrings muscles with relative sparing of their central part. This was best appreciated in the third decade of life, but could also be identified both in the younger patient with minimal MRI changes and in the oldest patient, despite her more severe and diffuse muscle involvement. This report suggests that muscle MRI could be used as an additional tool to establish the pattern and the degree of muscle involvement in patients with Bethlem myopathy. Further studies in a larger cohort are needed to evaluate the specificity of these findings.


Assuntos
Colágeno Tipo VI/genética , Imageamento por Ressonância Magnética , Músculo Esquelético/patologia , Doenças Musculares/genética , Doenças Musculares/patologia , Adulto , Idoso , Biópsia , Criança , Análise Mutacional de DNA , Feminino , Humanos , Mutação Puntual/genética
19.
Semin Pediatr Neurol ; 9(2): 120-31, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12138996

RESUMO

The number of new syndromes, loci, and genes responsible for CMD forms has dramatically increased in the last few years, and it has become increasingly evident that the classification of the different forms of CMD is a difficult task. A recent classification separated the forms of CMD that have been mapped (CMD diseases) from the ones with clearly defined clinical and pathologic features that have not been mapped yet (CMD syndromes). Eight CMD forms have been mapped up to now, and the genes responsible for three of them have been identified. This review describes an update of clinical, pathologic, and genetic findings in the different CMD forms.


Assuntos
Distrofias Musculares/congênito , Distrofias Musculares/genética , Encéfalo/anormalidades , Anormalidades do Olho , Expressão Gênica/genética , Humanos , Laminina/genética , Laminina/metabolismo , Imageamento por Ressonância Magnética , Biologia Molecular/métodos , Músculo Esquelético/patologia , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia
20.
Neuromuscul Disord ; 24(3): 250-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24342283

RESUMO

Several human and animal myopathies, such as malignant hyperthermia (MH), central core disease and equine recurrent exertional rhabdomyolysis (RER) are confirmed or thought to be associated with dysfunction of skeletal muscle calcium regulation. For some patients in whom the genetic cause is unknown, or when mutational analysis reveals genetic variants with unclear pathogenicity, defects are further studied through use of muscle histopathology and in vitro contraction tests, the latter in particular, when assessing responses to ryanodine receptor agonists, such as caffeine. However, since muscle biopsy is not always suitable, researchers have used cultured cells to model these diseases, by examining calcium regulation in myotubes derived from skin, following forced expression of muscle-specific transcription factors. Here we describe a novel adenoviral vector that we used to express equine MyoD in dermal fibroblasts. In permissive conditions, transduced equine and human fibroblasts differentiated into multinucleated myotubes. We demonstrate that these cells have a functional excitation-calcium release mechanism and, similarly to primary muscle-derived myotubes, respond in a dose-dependent manner to increasing concentrations of caffeine. MyoD-induced conversion of equine skin-derived fibroblasts offers an attractive method for evaluating calcium homeostasis defects in vitro without the need for invasive muscle biopsy.


Assuntos
Adenoviridae/genética , Cafeína/farmacologia , Derme/citologia , Fibroblastos/metabolismo , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteína MyoD/metabolismo , Animais , Células Cultivadas , Fibroblastos/citologia , Cavalos , Humanos , Masculino , Fibras Musculares Esqueléticas/citologia , Proteína MyoD/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA