Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Med ; 4(4): 245-251.e3, 2023 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-36905929

RESUMO

BACKGROUND: Utrophin, a dystrophin homolog, is consistently upregulated in muscles of patients with Duchenne muscular dystrophy (DMD) and is believed to partially compensate for the lack of dystrophin in dystrophic muscle. Even though several animal studies support the idea that utrophin can modulate DMD disease severity, human clinical data are scarce. METHODS: We describe a patient with the largest reported in-frame deletion in the DMD gene, including exons 10-60 and thus encompassing the entire rod domain. FINDINGS: The patient presented with an unusually early and severe progressive weakness, initially suggesting congenital muscular dystrophy. Immunostaining of his muscle biopsy showed that the mutant protein was able to localize at the sarcolemma and stabilize the dystrophin-associated complex. Strikingly, utrophin protein was absent from the sarcolemmal membrane despite the upregulation of utrophin mRNA. CONCLUSIONS: Our results suggest that the internally deleted and dysfunctional dystrophin lacking the entire rod domain may exert a dominant-negative effect by preventing upregulated utrophin protein from reaching the sarcolemmal membrane and thus blocking its partial rescue of muscle function. This unique case may set a lower size limit for similar constructs in potential gene therapy approaches. FUNDING: This work was supported by a grant from MDA USA (MDA3896) and by grant number R01AR051999 from NIAMS/NIH to C.G.B.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Humanos , Distrofina/genética , Distrofina/metabolismo , Utrofina/genética , Utrofina/metabolismo , Utrofina/uso terapêutico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patologia , Músculos/metabolismo , Músculos/patologia , Sarcolema/metabolismo , Sarcolema/patologia
2.
J Agric Food Chem ; 65(51): 11212-11219, 2017 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-29199426

RESUMO

Anchovy (Engraulis japonicus) meat (AM) has been shown to promote nonheme iron absorption via a ferric oxyhydroxide nanoparticle (FeONP)-mediated mechanism. Here, formulation modifications of an egg-white-based AIN-93G diet with AM fractions resulted hemoglobin regeneration efficiencies in anemic rats following an order control (23.69 ± 3.99%) < ferrous-sulfate-replacement of ferric citrate (39.89 ± 2.97%) ≈ dehemeed-AM-protein-replacement of egg white (45.88 ± 4.76%) ≈ AM-lipid-replacement of soybean oil (43.14 ± 3.48%) ≈ chondroitin-sulfate-replacement of ∼2.5% corn starch (39.92 ± 1.88%) < l-α-phosphatidylcholine-replacement of ∼29% soybean oil (53.42 ± 2.04%), with nanosized iron enriched in proximal-small-intestinal contents by these AM fractions. The calcein-fluorescence-quenching assay in polarized Caco-2 cells revealed good iron absorption from FeONPs coated with AM peptides, l-α-phosphatidylcholine, l-α-lysophosphatidylcholine, and chondroitin sulfate, with the latter two disfavoring endocytosis thereby inducing relatively weaker iron absorption. These results suggest peptides, phospholipids, and mucopolysaccharides released during AM digestion are key factors promoting nonheme iron absorption.


Assuntos
Anemia/dietoterapia , Ferro/metabolismo , Carne/análise , Animais , Células CACO-2 , Digestão , Feminino , Compostos Férricos/metabolismo , Peixes , Heme/metabolismo , Hemoglobinas/metabolismo , Humanos , Nanopartículas/metabolismo , Ratos , Ratos Sprague-Dawley
3.
Hum Mol Genet ; 26(12): 2207-2217, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28419360

RESUMO

Collagen prolyl 4-hydroxylases (C-P4Hs) play a central role in the formation and stabilization of the triple helical domain of collagens. P4HA1 encodes the catalytic α(I) subunit of the main C-P4H isoenzyme (C-P4H-I). We now report human bi-allelic P4HA1 mutations in a family with a congenital-onset disorder of connective tissue, manifesting as early-onset joint hypermobility, joint contractures, muscle weakness and bone dysplasia as well as high myopia, with evidence of clinical improvement of motor function over time in the surviving patient. Similar to P4ha1 null mice, which die prenatally, the muscle tissue from P1 and P2 was found to have reduced collagen IV immunoreactivity at the muscle basement membrane. Patients were compound heterozygous for frameshift and splice site mutations leading to reduced, but not absent, P4HA1 protein level and C-P4H activity in dermal fibroblasts compared to age-matched control samples. Differential scanning calorimetry revealed reduced thermal stability of collagen in patient-derived dermal fibroblasts versus age-matched control samples. Mutations affecting the family of C-P4Hs, and in particular C-P4H-I, should be considered in patients presenting with congenital connective tissue/myopathy overlap disorders with joint hypermobility, contractures, mild skeletal dysplasia and high myopia.


Assuntos
Pró-Colágeno-Prolina Dioxigenase/genética , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Prolil Hidroxilases/genética , Animais , Membrana Basal/metabolismo , Osso e Ossos/metabolismo , Criança , Colágeno Tipo IV/genética , Tecido Conjuntivo , Humanos , Masculino , Camundongos , Camundongos Knockout , Músculos/metabolismo , Mutação , Osteocondrodisplasias/genética , Prolil Hidroxilases/metabolismo , Tendões/metabolismo
4.
JAMA Neurol ; 72(6): 689-98, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25938801

RESUMO

IMPORTANCE: New genomic strategies can now be applied to identify a diagnosis in patients and families with previously undiagnosed rare genetic conditions. The large family evaluated in the present study was described in 1966 and now expands the phenotype of a known neuromuscular gene. OBJECTIVE: To determine the genetic cause of a slowly progressive, autosomal dominant, scapuloperoneal neuromuscular disorder by using linkage and exome sequencing. DESIGN, SETTING, AND PARTICIPANTS: Fourteen affected individuals in a 6-generation family with a progressive scapuloperoneal disorder were evaluated. Participants were examined at pediatric, neuromuscular, and research clinics from March 1, 2005, to May 31, 2014. Exome and linkage were performed in genetics laboratories of research institutions. MAIN OUTCOMES AND MEASURES: Examination and evaluation by magnetic resonance imaging, ultrasonography, electrodiagnostic studies, and muscle biopsies (n = 3). Genetic analysis included linkage analysis (n = 17) with exome sequencing (n = 7). RESULTS: Clinical findings included progressive muscle weakness in an initially scapuloperoneal and distal distribution, including wrist extensor weakness, finger and foot drop, scapular winging, mild facial weakness, Achilles tendon contractures, and diminished or absent deep tendon reflexes. Both age at onset and progression of the disease showed clinical variability within the family. Muscle biopsy specimens demonstrated type I fiber atrophy and trabeculated fibers without nemaline rods. Analysis of exome sequences within the linkage region (4.8 megabases) revealed missense mutation c.591C>A p.Glu197Asp in a highly conserved residue in exon 4 of ACTA1. The mutation cosegregated with disease in all tested individuals and was not present in unaffected individuals. CONCLUSIONS AND RELEVANCE: This family defines a new scapuloperoneal phenotype associated with an ACTA1 mutation. A highly conserved protein, ACTA1 is implicated in multiple muscle diseases, including nemaline myopathy, actin aggregate myopathy, fiber-type disproportion, and rod-core myopathy. To our knowledge, mutations in Glu197 have not been reported previously. This residue is highly conserved and located in an exposed position in the protein; the mutation affects the intermolecular and intramolecular electrostatic interactions as shown by structural modeling. The mutation in this residue does not appear to lead to rod formation or actin accumulation in vitro or in vivo, suggesting a different molecular mechanism from that of other ACTA1 diseases.


Assuntos
Actinas/genética , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/fisiopatologia , Adulto , Idade de Início , Criança , Progressão da Doença , Exoma/genética , Ligação Genética , Humanos , Masculino , Distrofia Muscular de Emery-Dreifuss/patologia , Mutação de Sentido Incorreto/genética , Miopatias da Nemalina , Linhagem , Fenótipo
5.
J Vis Exp ; (95): 52307, 2015 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-25651101

RESUMO

The generation of patient-specific cell lines represents an invaluable tool for diagnostic or translational research, and these cells can be collected from skin or muscle biopsy tissue available during the patient's diagnostic workup. In this protocol, we describe a technique for live cell isolation from small amounts of muscle or skin tissue for primary cell culture. Additionally, we provide a technique for the immortalization of myogenic cell lines and fibroblast cell lines from primary cells. Once cell lines are immortalized, substantial expansion of patient-derived cells can be achieved. Immortalized cells are amenable to many downstream applications, including drug screening and in vitro correction of the genetic mutation. Altogether, these protocols provide a reliable tool to generate and preserve patient-derived cells for downstream applications.


Assuntos
Técnicas Citológicas/métodos , Fibroblastos/citologia , Técnicas de Diagnóstico Molecular/métodos , Músculos/citologia , Pele/citologia , Biópsia/métodos , Linhagem Celular , Separação Celular/métodos , Humanos
6.
Hum Mol Genet ; 23(9): 2353-63, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24334769

RESUMO

Bethlem myopathy (BM) [MIM 158810] is a slowly progressive muscle disease characterized by contractures and proximal weakness, which can be caused by mutations in one of the collagen VI genes (COL6A1, COL6A2 and COL6A3). However, there may be additional causal genes to identify as in ∼50% of BM cases no mutations in the COL6 genes are identified. In a cohort of -24 patients with a BM-like phenotype, we first sequenced 12 candidate genes based on their function, including genes for known binding partners of collagen VI, and those enzymes involved in its correct post-translational modification, assembly and secretion. Proceeding to whole-exome sequencing (WES), we identified mutations in the COL12A1 gene, a member of the FACIT collagens (fibril-associated collagens with interrupted triple helices) in five individuals from two families. Both families showed dominant inheritance with a clinical phenotype resembling classical BM. Family 1 had a single-base substitution that led to the replacement of one glycine residue in the triple-helical domain, breaking the Gly-X-Y repeating pattern, and Family 2 had a missense mutation, which created a mutant protein with an unpaired cysteine residue. Abnormality at the protein level was confirmed in both families by the intracellular retention of collagen XII in patient dermal fibroblasts. The mutation in Family 2 leads to the up-regulation of genes associated with the unfolded protein response (UPR) pathway and swollen, dysmorphic rough-ER. We conclude that the spectrum of causative genes in extracellular matrix (ECM)-related myopathies be extended to include COL12A1.


Assuntos
Colágeno Tipo XII/genética , Colágeno/genética , Matriz Extracelular/metabolismo , Doenças Musculares/genética , Doenças Musculares/patologia , Adolescente , Adulto , Criança , Colágeno Tipo VI/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Adulto Jovem
7.
J Biol Chem ; 285(13): 10005-10015, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20106987

RESUMO

Ullrich congenital muscular dystrophy (UCMD) is a disabling and life-threatening disorder resulting from either recessive or dominant mutations in genes encoding collagen VI. Although the majority of the recessive UCMD cases have frameshift or nonsense mutations in COL6A1, COL6A2, or COL6A3, recessive structural mutations in the COL6A2 C-globular region are emerging also. However, the underlying molecular mechanisms have remained elusive. Here we identified a homozygous COL6A2 E624K mutation (C1 subdomain) and a homozygous COL6A2 R876S mutation (C2 subdomain) in two UCMD patients. The consequences of the mutations were investigated using fibroblasts from patients and cells stably transfected with the mutant constructs. In contrast to expectations based on the clinical severity of these two patients, secretion and assembly of collagen VI were moderately affected by the E624K mutation but severely impaired by the R876S substitution. The E624K substitution altered the electrostatic potential of the region surrounding the metal ion-dependent adhesion site, resulting in a collagen VI network containing thick fibrils and spots with densely packed microfibrils. The R876S mutation prevented the chain from assembling into triple-helical collagen VI molecules. The minute amount of collagen VI secreted by the R876S fibroblasts was solely composed of a faster migrating chain corresponding to the C2a splice variant with an alternative C2 subdomain. In transfected cells, the C2a splice variant was able to assemble into short microfibrils. Together, the results suggest that the C2a splice variant may functionally compensate for the loss of the normal COL6A2 chain when mutations occur in the C2 subdomain.


Assuntos
Processamento Alternativo , Colágeno Tipo VI/genética , Genes Recessivos , Distrofias Musculares/congênito , Distrofias Musculares/genética , Mutação de Sentido Incorreto , Adulto , Sequência de Aminoácidos , Biópsia , Criança , Colágeno/química , Feminino , Fibroblastos/metabolismo , Homozigoto , Humanos , Íons , Masculino , Dados de Sequência Molecular , Homologia de Sequência de Aminoácidos
8.
Brain ; 132(Pt 2): 452-64, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19181672

RESUMO

We recently identified the X-chromosomal four and a half LIM domain gene FHL1 as the causative gene for reducing body myopathy, a disorder characterized by progressive weakness and intracytoplasmic aggregates in muscle that exert reducing activity on menadione nitro-blue-tetrazolium (NBT). The mutations detected in FHL1 affected highly conserved zinc coordinating residues within the second LIM domain and lead to the formation of aggregates when transfected into cells. Our aim was to define the clinical and morphological phenotype of this myopathy and to assess the mutational spectrum of FHL1 mutations in reducing body myopathy in a larger cohort of patients. Patients were ascertained via the detection of reducing bodies in muscle biopsy sections stained with menadione-NBT followed by clinical, histological, ultrastructural and molecular genetic analysis. A total of 11 patients from nine families were included in this study, including seven sporadic patients with early childhood onset disease and four familial cases with later onset. Weakness in all patients was progressive, sometimes rapidly so. Respiratory failure was common and scoliosis and spinal rigidity were significant in some of the patients. Analysis of muscle biopsies confirmed the presence of aggregates of FHL1 positive material in all biopsies. In two patients in whom sequential biopsies were available the aggregate load in muscle sections appeared to increase over time. Ultrastructural analysis revealed that cytoplasmic bodies were regularly seen in conjunction with the reducing bodies. The mutations detected were exclusive to the second LIM domain of FHL1 and were found in both sporadic as well as familial cases of reducing body myopathy. Six of the nine mutations affected the crucial zinc coordinating residue histidine 123. All mutations in this residue were de novo and were associated with a severe clinical course, in particular in one male patient (H123Q). Mutations in the zinc coordinating residue cysteine 153 were associated with a milder phenotype and were seen in the familial cases in which the boys were still more severely affected compared to their mothers. We expect the mild end of the spectrum to significantly expand in the future. On the severe end of the spectrum we define reducing body myopathy as a progressive disease with early, but not necessarily congenital onset, distinguishing this condition from the classic essentially non-progressive congenital myopathies.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Musculares/genética , Doenças Musculares/genética , Mutação de Sentido Incorreto , Adulto , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença , Humanos , Lactente , Proteínas com Domínio LIM , Masculino , Microscopia Imunoeletrônica , Músculo Esquelético/patologia , Doenças Musculares/patologia , Linhagem
9.
Muscle Nerve ; 38(3): 1184-91, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18720506

RESUMO

Ullrich disease (congenital muscular dystrophy type Ullrich, UCMD) is a severe congenital disorder of muscle caused by recessive and dominant mutations in the three genes that encode the alpha-chains of collagen type VI. Little is known about the early pathogenesis of this myopathy. The aim of this study was to investigate early histological changes in muscle of patients with molecularly confirmed UCMD. Muscle biopsies were analyzed from 8 UCMD patients ranging in age from 6 to 30 months. Type I fiber atrophy and predominance were seen early, together with a widening of the fiber diameter spectrum, whereas no dystrophic features were apparent. A subpopulation of more severely atrophic type I fibers was apparent subsequently, including one biopsy that fulfilled the formal diagnostic criteria of histopathological fiber type disproportion (FTD). Thus, early in the disease, UCMD presents as a non-dystrophic myopathy with predominant fiber atrophy. Collagen VI mutations also qualify as a cause of fiber type disproportion.


Assuntos
Fibras Musculares Esqueléticas/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/patologia , Distrofia Miotônica/complicações , Adenosina Trifosfatases/metabolismo , Biópsia/métodos , Pré-Escolar , Colágeno Tipo VI/metabolismo , Feminino , Humanos , Indóis , Lactente , Laminina/metabolismo , Masculino , Fibras Musculares Esqueléticas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Distrofia Miotônica/patologia
10.
Neuromuscul Disord ; 17(1): 28-32, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17134899

RESUMO

Dynamin 2 has recently been recognized as a causative gene for the autosomal dominant form of centronuclear myopathy (dominant centronuclear myopathy). Here we report an affected father and daughter with dynamin 2 related AD CNM with predominantly distal onset of weakness. In addition to the diagnostic central location of myonuclei the muscle biopsy also showed core-like structures. Muscle MRI in the lower leg revealed prominent involvement of the soleus, but also of the gastrocnemius and the tibialis anterior whereas in the thigh there was a consistent pattern of selective involvement of adductor longus, semimembranosus, biceps femoris, rectus femoris, and vastus intermedius with relative sparing of vastus lateralis and medialis, sartorius, gracilis, and partly of the semitendinosus. These characteristic findings on muscle MRI confirm similar findings reported for CT imaging in dynamin 2 related dominant centronuclear myopathy and may help to differentiate this disorder from central core disease and other myopathies.


Assuntos
Dinamina II/genética , Músculo Esquelético/patologia , Miopatias Congênitas Estruturais/genética , Miopatias Congênitas Estruturais/patologia , Adulto , Dinamina II/metabolismo , Saúde da Família , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Mutação
11.
Am J Med Genet A ; 132A(3): 296-301, 2005 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-15690374

RESUMO

Ullrich congenital muscular dystrophy (UCMD) is caused by mutations in the three genes coding for the alpha chains of collagen VI and characterized by generalized muscle weakness, striking hypermobility of distal joints in conjunction with variable contractures of more proximal joints, and normal intellectual development. The diagnosis is supported by abnormal immunoreactivity for collagen VI on muscle biopsies. As patients with UCMD show clinical characteristics typical of classical disorders of connective tissue such as Ehlers-Danlos syndromes (EDS), we investigated the ultrastructure of skin biopsy samples from patients with UCMD (n=5). Electron microscopy of skin biopsies revealed ultrastructural abnormalities in all cases, including alterations of collagen fibril morphology (variation in size and composite fibers) and increase in ground substance, which resemble those seen in patients with EDS. Our findings suggest that there is a true connective tissue component as part of the phenotypic spectrum of UCMD and that there is considerable clinical as well as morphological overlap between UCMD and classic connective tissue disorders.


Assuntos
Tecido Conjuntivo/anormalidades , Síndrome de Ehlers-Danlos/diagnóstico , Distrofias Musculares/diagnóstico , Pele/patologia , Adolescente , Criança , Diagnóstico Diferencial , Humanos , Microscopia Eletrônica , Distrofias Musculares/congênito , Pele/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA