Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Hum Gene Ther Methods ; 27(5): 174-186, 2016 10.
Article in English | MEDLINE | ID: mdl-27477497

ABSTRACT

Duchenne muscular dystrophy (DMD) is a severe, genetic muscle disease caused by the absence of the sarcolemmal protein dystrophin. Gene replacement therapy is considered a potential strategy for the treatment of DMD, aiming to restore the missing protein. Although the elements of the dystrophin molecule have been identified and studies in transgenic mdx mice have explored the importance of a number of these structural domains, the resulting modified dystrophin protein products that have been developed so far are only partially characterized in relation to their structure and function in vivo. To optimize a dystrophin cDNA construct for therapeutic application we designed and produced four human minidystrophins within the packaging capacity of lentiviral vectors. Two novel minidystrophins retained the centrally located neuronal nitric oxide synthase (nNOS)-anchoring domain in order to achieve sarcolemmal nNOS restoration, which is lost in most internally deleted dystrophin constructs. Functionality of the resulting truncated dystrophin proteins was investigated in muscle of adult dystrophin-deficient mdx mice followed by a battery of detailed immunohistochemical and morphometric tests. This initial assessment aimed to determine the overall suitability of various constructs for cloning into lentiviral vectors for ex vivo gene delivery to stem cells for future preclinical studies.


Subject(s)
Dystrophin/genetics , Genetic Therapy , Muscular Dystrophy, Duchenne/therapy , Nitric Oxide Synthase Type I/genetics , Animals , DNA, Complementary/genetics , DNA, Complementary/therapeutic use , Dystrophin/therapeutic use , Gene Expression , Genetic Vectors/therapeutic use , Humans , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Nitric Oxide Synthase Type I/biosynthesis
2.
Biomolecules ; 5(4): 2758-81, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26501342

ABSTRACT

Glutamine-fructose-6-phosphate transaminase 1 (GFPT1) is the first enzyme of the hexosamine biosynthetic pathway. It transfers an amino group from glutamine to fructose-6-phosphate to yield glucosamine-6-phosphate, thus providing the precursor for uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) synthesis. UDP-GlcNAc is an essential substrate for all mammalian glycosylation biosynthetic pathways and N-glycan branching is especially sensitive to alterations in the concentration of this sugar nucleotide. It has been reported that GFPT1 mutations lead to a distinct sub-class of congenital myasthenic syndromes (CMS) termed "limb-girdle CMS with tubular aggregates". CMS are hereditary neuromuscular transmission disorders in which neuromuscular junctions are impaired. To investigate whether alterations in protein glycosylation at the neuromuscular junction might be involved in this impairment, we have employed mass spectrometric strategies to study the N-glycomes of myoblasts and myotubes derived from two healthy controls, three GFPT1 patients, and four patients with other muscular diseases, namely CMS caused by mutations in DOK7, myopathy caused by mutations in MTND5, limb girdle muscular dystrophy type 2A (LGMD2A), and Pompe disease. A comparison of the relative abundances of bi-, tri-, and tetra-antennary N-glycans in each of the cell preparations revealed that all samples exhibited broadly similar levels of branching. Moreover, although some differences were observed in the relative abundances of some of the N-glycan constituents, these variations were modest and were not confined to the GFPT1 samples. Therefore, GFPT1 mutations in CMS patients do not appear to compromise global N-glycosylation in muscle cells.


Subject(s)
Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/genetics , Myasthenic Syndromes, Congenital/metabolism , Myoblasts/metabolism , Protein Processing, Post-Translational , Adolescent , Adult , Case-Control Studies , Cells, Cultured , Electron Transport Complex I/genetics , Female , Glycosylation , Humans , Male , Middle Aged , Mitochondrial Proteins/genetics , Muscle Proteins/genetics , Myasthenic Syndromes, Congenital/genetics
3.
Genesis ; 52(7): 687-94, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24700590

ABSTRACT

Nipbl (Scc2) and Mau2 (Scc4) encode evolutionary conserved proteins that play a vital role for loading the cohesin complex onto chromosomes, thereby ensuring accurate chromosome segregation during cell division. While mutations in human NIPBL are known to cause the developmental disorder Cornelia de Lange syndrome, the functions of Nipbl and Mau2 in mammalian development are poorly defined. Here we generated conditional alleles for both genes in mice and show that neural crest cell-specific inactivation of Nipbl or Mau2 strongly affects craniofacial development. Surprisingly, the early phase of neural crest cell proliferation and migration is only moderately affected in these mutants. Moreover, we found that Mau2 single homozygous mutants exhibited a more severe craniofacial phenotype when compared to that of Nipbl;Mau2 double homozygous mutants. This raises the possibility that the Mau2/Nipbl protein interaction is not only required for cohesin loading, but may also be required to restrict the level of Nipbl involved in regulating gene expression. Together, the data suggest that proliferating neural crest cells tolerate a substantial reduction of cohesin loading proteins and we propose that the successive decrease of cohesin loading proteins in neural crest cells may alter developmental gene regulation in a highly dynamic manner.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , Craniofacial Abnormalities/genetics , Neural Crest/metabolism , Transcription Factors/genetics , Animals , Cell Cycle Proteins , Chromosomal Proteins, Non-Histone/metabolism , Craniofacial Abnormalities/embryology , DNA-Binding Proteins , Female , Male , Mice , Transcription Factors/metabolism
4.
PLoS Curr ; 52013 May 21.
Article in English | MEDLINE | ID: mdl-23788081

ABSTRACT

Advances in the molecular characterisation of genetic muscle disease has been rapid, as demonstrated by a recent analysis of these conditions in the north of England by Norwood et al (2009), in which a genetic diagnosis was achieved for 75.7% of patients. However, there remain many patients with suspected genetic muscle disease in who a diagnosis is not obtained, often despite considerable diagnostic effort, and these patients are now being considered for the application of new technologies such as next generation sequencing. This study aimed to provide an in-depth phenotype analysis of undiagnosed patients referred to the Northern region muscle clinic with suspected genetic muscle disease, with the intention of gaining insight into these conditions, identifing cases with a shared phenotype who may be amenable to collective diagnostic testing or research, and evaluating the strengths and limitations of our current diagnostic strategy. We used two approaches: a review of clinical findings in patients with undiagnosed muscle disease, and a hierarchical cluster analysis to provide an unbiased interpretation of the phenotype data. These joint approaches identified a correlation of phenotypic features according to the age of disease onset and also delineated several interesting groups of patients, as well as highlighting areas of frequent diagnostic difficulty that could benefit from the use of new high-throughput diagnostic techniques. Correspondence to: anna.sarkozy@ncl.ac.uk.

5.
Stem Cells Dev ; 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23786351

ABSTRACT

With an incidence of ~1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harbouring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ~30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.

6.
Hum Mutat ; 34(8): 1111-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23606453

ABSTRACT

Limb girdle muscular dystrophy type 2L or anoctaminopathy is a condition mainly characterized by adult onset proximal lower limb muscular weakness and raised CK values, due to recessive ANO5 gene mutations. An exon 5 founder mutation (c.191dupA) has been identified in most of the British and German LGMD2L patients so far reported. We aimed to further investigate the prevalence and spectrum of ANO5 gene mutations and related clinical phenotypes, by screening 205 undiagnosed patients referred to our molecular service with a clinical suspicion of anoctaminopathy. A total of 42 unrelated patients had two ANO5 mutations (21%), whereas 14 carried a single change. We identified 34 pathogenic changes, 15 of which are novel. The c.191dupA mutation represents 61% of mutated alleles and appears to be less prevalent in non-Northern European populations. Retrospective clinical analysis corroborates the prevalently proximal lower limb phenotype, the male predominance and absence of major cardiac or respiratory involvement. Identification of cases with isolated hyperCKaemia and very late symptomatic male and female subjects confirms the extension of the phenotypic spectrum of the disease. Anoctaminopathy appears to be one of the most common adult muscular dystrophies in Northern Europe, with a prevalence of about 20%-25% in unselected undiagnosed cases.


Subject(s)
Chloride Channels/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Adult , Aged , Anoctamins , Chloride Channels/metabolism , Europe/epidemiology , Female , Genetic Variation , Humans , Male , Middle Aged , Muscular Dystrophies, Limb-Girdle/epidemiology , Muscular Dystrophies, Limb-Girdle/metabolism , Phenotype , Prevalence , Retrospective Studies , Sex Factors
7.
PLoS One ; 8(2): e57260, 2013.
Article in English | MEDLINE | ID: mdl-23437355

ABSTRACT

Beta-blockers are used to treat acquired heart failure in adults, though their role in early muscular dystrophy cardiomyopathy is unclear. We treated 2 different dystrophic mouse models which have an associated cardiomyopathy (mdx: model for Duchenne Muscular Dystrophy, and Sgcd-/-: model for limb girdle muscular dystrophy type 2F) and wild type controls (C57 Bl10) with the beta blocker metoprolol or placebo for 8 weeks at an early stage in the development of the cardiomyopathy. Left and right ventricular function was assessed with cardiac magnetic resonance imaging (MRI) and in-vivo myocardial calcium influx with manganese enhanced MRI. In the mdx mice at baseline there was reduced stroke volume, cardiac index, and end-diastolic volume with preserved left ventricular ejection fraction. These abnormalities were no longer evident after treatment with beta-blockers. Right ventricular ejection fraction was reduced and right ventricular end-systolic volume increased in the mdx mice. With metoprolol there was an increase in right ventricular end-diastolic and end-systolic volumes. Left and right ventricular function was normal in the Sgcd-/- mice. Metroprolol had no significant effects on left and right ventricular function in these mice, though heart/body weight ratios increased after treatment. In-vivo myocardial calcium influx with MEMRI was significantly elevated in both models, though metoprolol had no significant effects on either. In conclusion, metoprolol treatment at an early stage in the development of cardiomyopathy has deleterious effects on right ventricular function in mdx mice and in both models no effect on increased in-vivo calcium influx. This suggests that clinical trials need to carefully monitor not just left ventricular function but also right ventricular function and other aspects of myocardial metabolism.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Calcium/metabolism , Cardiomyopathies/drug therapy , Metoprolol/pharmacology , Muscular Dystrophies/drug therapy , Ventricular Function, Left/drug effects , Ventricular Function, Right/drug effects , Animals , Body Weight/drug effects , Calcium/agonists , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Ion Transport/drug effects , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Organ Size/drug effects , Sarcoglycans/deficiency , Sarcoglycans/genetics , Stroke Volume/drug effects
8.
J Cardiovasc Magn Reson ; 15: 4, 2013 Jan 16.
Article in English | MEDLINE | ID: mdl-23324314

ABSTRACT

BACKGROUND: Manganese-enhanced cardiovascular magnetic resonance (MECMR) can non-invasively assess myocardial calcium influx, and calcium levels are known to be elevated in muscular dystrophy cardiomyopathy based on cellular studies. METHODS: Left ventricular functional studies and MECMR were performed in mdx mice (model of Duchenne muscular dystrophy, 24 and 40 weeks) and Sgcd -/- mice (limb girdle muscular dystrophy 2 F, 16 and 32 weeks), compared to wild type controls (C57Bl/10, WT). RESULTS: Both models had left ventricular hypertrophy at the later age compared to WT, though the mdx mice had reduced stroke volumes and the Sgcd -/- mice increased heart rate and cardiac index. Especially at the younger ages, MECMR was significantly elevated in both models (both P < 0.05 versus WT). The L-type calcium channel inhibitor diltiazem (5 mg/kg i.p.) significantly reduced MECMR in the mdx mice (P < 0.01), though only with a higher dose (10 mg/kg i.p.) in the Sgcd -/- mice (P < 0.05). As the Sgcd -/- mice had increased heart rates, to determine the role of heart rate in MECMR we studied the hyperpolarization-activated cyclic nucleotide-gated channel inhibitor ZD 7288 which selectively reduces heart rate. This reduced heart rate and MECMR in all mouse groups. However, when looking at the time course of reduction of MECMR in the Sgcd -/- mice at up to 5 minutes of the manganese infusion when heart rates were matched to the WT mice, MECMR was still significantly elevated in the Sgcd -/- mice (P < 0.01) indicating that heart rate alone could not account for all the increased MECMR. CONCLUSIONS: Despite both mouse models exhibiting increased in-vivo calcium influx at an early stage in the development of the cardiomyopathy before left ventricular hypertrophy, there are distinct phenotypical differences between the 2 models in terms of heart rates, hemodynamics and responses to calcium channel inhibitors.


Subject(s)
Calcium Signaling , Cardiomyopathies/metabolism , Muscular Dystrophy, Duchenne/metabolism , Myocardium/metabolism , Ventricular Function, Left , Age Factors , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Chlorides , Contrast Media , Disease Models, Animal , Disease Progression , Genotype , Heart Rate , Hypertrophy, Left Ventricular/metabolism , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Magnetic Resonance Imaging , Male , Manganese Compounds , Mice , Mice, Inbred mdx , Mice, Knockout , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Myocardium/pathology , Phenotype , Sarcoglycans/deficiency , Sarcoglycans/genetics , Stroke Volume , Time Factors , Ventricular Function, Left/drug effects
9.
Hum Mol Genet ; 21(8): 1706-24, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22199023

ABSTRACT

Ariel is a mouse mutant that suffers from skeletal muscle myofibrillar degeneration due to the rapid accumulation of large intracellular protein aggregates. This fulminant disease is caused by an ENU-induced recessive mutation resulting in an L342Q change within the motor domain of the skeletal muscle myosin protein MYH4 (MyHC IIb). Although normal at birth, homozygous mice develop hindlimb paralysis from Day 13, consistent with the timing of the switch from developmental to adult myosin isoforms in mice. The mutated myosin (MYH4(L342Q)) is an aggregate-prone protein. Notwithstanding the speed of the process, biochemical analysis of purified aggregates showed the presence of proteins typically found in human myofibrillar myopathies, suggesting that the genesis of ariel aggregates follows a pathogenic pathway shared with other conformational protein diseases of skeletal muscle. In contrast, heterozygous mice are overtly and histologically indistinguishable from control mice. MYH4(L342Q) is present in muscles from heterozygous mice at only 7% of the levels of the wild-type protein, resulting in a small but significant increase in force production in isolated single fibres and indicating that elimination of the mutant protein in heterozygotes prevents the pathological changes observed in homozygotes. Recapitulation of the L342Q change in the functional equivalent of mouse MYH4 in human muscles, MYH1, results in a more aggregate-prone protein.


Subject(s)
Muscular Diseases/genetics , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/genetics , Amino Acid Sequence , Animals , Genes, Recessive , Heterozygote , Homozygote , Humans , Mice , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Mutation , Myofibrils/ultrastructure , Myosin Heavy Chains/metabolism , Protein Conformation , Protein Structure, Tertiary , Transcription, Genetic
10.
Hum Mol Genet ; 20(24): 4879-90, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-21926082

ABSTRACT

Fukutin and fukutin-related protein (FKRP) are involved in the glycosylation of α-dystroglycan, a key receptor for basement membrane proteins. Aberrant α-dystroglycan glycosylation leads to a broad spectrum of disorders, ranging from limb girdle muscular dystrophy to Walker-Warburg syndrome. This is the first study investigating a role of fukutin and FKRP-mediated glycosylation in angiogenesis. Transgenic zebrafish expressing enhanced green fluorescent protein in blood vessels were treated with morpholino antisense oligonucleotides that blocked the expression of fukutin, FKRP and dystroglycan. All morphant fish showed muscle damage and vascular abnormalities at day 1 post-fertilization. Intersegmental vessels of somites failed to reach the dorsal longitudinal anastomosis and in more severe phenotypes retracted further or were in some cases even completely missing. In contrast, the eye vasculature was distorted in both fukutin and FKRP morphants, but not in dystroglycan morphants or control fish. The eye size was also smaller in the fukutin and FKRP morphants when compared with dystroglycan knockdown fish and controls. In general, the fukutin morphant fish had the most severe skeletal muscle and eye phenotype. Our findings suggest that fukutin and FKRP have functions that affect ocular development in zebrafish independently of dystroglycan. Despite anecdotal reports about vascular abnormalities in patients affected by dystroglycanopathies, the clinical relevance of such lesions remains unclear and should be subject to further review and investigations.


Subject(s)
Blood Vessels/abnormalities , Blood Vessels/embryology , Glycosyltransferases/deficiency , Zebrafish Proteins/deficiency , Zebrafish/embryology , Animals , Animals, Genetically Modified , Antibodies/immunology , Blood Vessels/drug effects , Blood Vessels/pathology , Dystroglycans/metabolism , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Eye/blood supply , Eye/drug effects , Eye/pathology , Glycosyltransferases/metabolism , Models, Animal , Morpholinos/pharmacology , Phalloidine/metabolism , Proto-Oncogene Protein c-fli-1 , Somites/abnormalities , Somites/blood supply , Somites/drug effects , Somites/embryology , Staining and Labeling , Zebrafish/genetics , Zebrafish Proteins/metabolism
11.
Am J Pathol ; 178(1): 273-83, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224064

ABSTRACT

The disease mechanisms underlying dystrophin-deficient muscular dystrophy are complex, involving not only muscle membrane fragility, but also dysregulated calcium homeostasis. Specifically, it has been proposed that calcium channels directly initiate a cascade of pathological events by allowing calcium ions to enter the cell. The objective of this study was to investigate the effect of chronically blocking calcium channels with the aminoglycoside antibiotic streptomycin from onset of disease in the mdx mouse model of Duchenne muscular dystrophy (DMD). Treatment in utero onwards delayed onset of dystrophic symptoms in the limb muscle of young mdx mice, but did not prevent degeneration and regeneration events occurring later in the disease course. Long-term treatment had a positive effect on limb muscle pathology, reduced fibrosis, increased sarcolemmal stability, and promoted muscle regeneration in older mice. However, streptomycin treatment did not show positive effects in diaphragm or heart muscle, and heart pathology was worsened. Thus, blocking calcium channels even before disease onset does not prevent dystrophy, making this an unlikely treatment for DMD. These findings highlight the importance of analyzing several time points throughout the life of the treated mice, as well as analyzing many tissues, to get a complete picture of treatment efficacy.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels/metabolism , Calcium/metabolism , Heart/drug effects , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/prevention & control , Animals , Diaphragm/drug effects , Diaphragm/pathology , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myocardium/pathology , Streptomycin/therapeutic use
12.
J Biol Chem ; 286(7): 5404-13, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21119217

ABSTRACT

Dysferlin (DYSF) is a type II transmembrane protein implicated in surface membrane repair of muscle. Mutations in dysferlin lead to Limb Girdle Muscular Dystrophy 2B (LGMD2B), Miyoshi Myopathy (MM), and Distal Myopathy with Anterior Tibialis onset (DMAT). The DYSF protein complex is not well understood, and only a few protein-binding partners have been identified thus far. To increase the set of interacting protein partners for DYSF we recovered a list of predicted interacting protein through a systems biology approach. The predictions are part of a "reverse-engineered" genome-wide human gene regulatory network obtained from experimental data by computational analysis. The reverse-engineering algorithm behind the analysis relates genes to each other based on changes in their expression patterns. DYSF and AHNAK were used to query the system and extract lists of potential interacting proteins. Among the 32 predictions the two genes share, we validated the physical interaction between DYSF protein with moesin (MSN) and polymerase I and transcript release factor (PTRF) in mouse heart lysate, thus identifying two novel Dysferlin-interacting proteins. Our strategy could be useful to clarify Dysferlin function in intracellular vesicles and its implication in muscle membrane resealing.


Subject(s)
Cell Membrane/metabolism , Gene Expression Regulation , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Algorithms , Animals , COS Cells , Cell Membrane/genetics , Chlorocebus aethiops , Distal Myopathies/genetics , Distal Myopathies/metabolism , Dysferlin , Humans , Membrane Proteins/genetics , Mice , Mice, Mutant Strains , Muscle Proteins/genetics , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism
13.
Eur J Heart Fail ; 12(11): 1163-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20675662

ABSTRACT

AIMS: Patients with mutations predisposing to cardiomyopathy often have routine assessments of left ventricular function. It is unclear whether asymptomatic mild cardiomyopathy should be treated with standard heart failure therapies. METHODS AND RESULTS: We tested the effect of metoprolol on cardiac haemodynamics and pathology in two animal models for muscular dystrophy and cardiomyopathy. Treatment started at an early stage in the development of the cardiomyopathy. Metoprolol was given orally (2.5 mg/kg/day) over 8 weeks to mdx mice (model for Duchenne muscular dystrophy) and δ-sarcoglycan-deficient (Sgcd(null)) mice (model for Limb girdle muscular dystrophy type 2F). In vivo pressure-volume loops, fibrosis, in vivo myocyte sarcolemmal injury, and ß-adrenergic receptor mRNA were assessed. In ß-blocked mdx mice, there was a beneficial reduction in afterload and restored contractility resulting in an increased stroke volume. In contrast, in Sgcd(null) mice, there was marked deterioration in haemodynamics (prolonged relaxation, Tau, and reduced stroke volume). Furthermore, challenging the ß-blocked Sgcd(null) mice with the ß-adrenergic agonist dobutamine led to markedly increased mortality. Patterns of sarcolemmal injury or ß-adrenergic receptor mRNA could not account for this, though the acute rise in markers of active relaxation suggested abnormally high levels of intracellular calcium. CONCLUSION: ß-Blockers may not necessarily be beneficial in all cardiomyopathies, even when given at an early stage of development. Clinical trials of ß-blockers in muscular dystrophy-associated cardiomyopathy may need to stratify patients by genotype.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/therapeutic use , Metoprolol/therapeutic use , Muscular Dystrophies, Limb-Girdle/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Adrenergic beta-1 Receptor Agonists/pharmacology , Adrenergic beta-1 Receptor Antagonists/administration & dosage , Animals , Disease Models, Animal , Dobutamine/pharmacology , Heart Failure/epidemiology , Hemodynamics/drug effects , Metoprolol/administration & dosage , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Inbred mdx , Muscular Dystrophies, Limb-Girdle/epidemiology , Muscular Dystrophies, Limb-Girdle/physiopathology , Muscular Dystrophy, Duchenne/epidemiology , Muscular Dystrophy, Duchenne/physiopathology , Papillary Muscles/pathology
14.
Muscle Nerve ; 41(2): 166-73, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20082313

ABSTRACT

Mutations in the dysferlin gene cause limb-girdle muscular dystrophy type 2B, Miyoshi myopathy, and distal anterior compartment myopathy. Dysferlin mainly localizes to the sarcolemma in mature skeletal muscle where it is implicated in membrane fusion and repair. In different forms of muscular dystrophy, a predominantly cytoplasmic localization of dysferlin can be observed in regenerating myofibers, but the subcellular compartment responsible for this labeling pattern is not yet known. We have previously demonstrated an association of dysferlin with the developing T-tubule system in vitro. To investigate the role of dysferlin in adult skeletal muscle regeneration, we studied dysferlin localization at high resolution in a rat model of regeneration and found that the subcellular labeling of dysferlin colocalizes with the developing T-tubule system. Furthermore, ultrastructural analysis of dysferlin-deficient muscle revealed primary T-tubule anomalies similar to those seen in caveolin-3-deficient muscle. These findings indicate that dysferlin is necessary for correct T-tubule formation, and dysferlin-deficient skeletal muscle is characterized by abnormally configured T-tubules.


Subject(s)
Membrane Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies/metabolism , Sarcolemma/metabolism , Animals , Biopsy , Cytoplasm/metabolism , Cytoplasm/pathology , Disease Models, Animal , Dysferlin , Female , Humans , Membrane Proteins/genetics , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Mutation/genetics , Rats , Rats, Wistar , Regeneration , Sarcolemma/pathology
15.
FASEB J ; 21(8): 1768-76, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17363620

ABSTRACT

The dysferlin gene is mutated in limb-girdle muscular dystrophy type 2B, Miyoshi myopathy, and distal anterior compartment myopathy. In mature skeletal muscle, dysferlin is located predominantly at the sarcolemma, where it plays a role in membrane fusion and repair. To investigate the role of dysferlin during early muscle differentiation, its localization was studied at high resolution in a muscle cell line. This demonstrated that dysferlin is not expressed at the plasmalemma of myotubes but mostly localizes to the T-tubule network. However, dysferlin translocated to the site of injury and toward the plasma membrane in a Ca2+-dependent fashion in response to a newly designed in vitro wounding assay. This reaction was specific to the full-length protein, as heterologously expressed deletion mutants of distinct C2 domains of dysferlin did not show this response. These results shed light on the dynamics of muscle membrane repair and are highly indicative of a specific role of dysferlin in this process in early myogenesis.


Subject(s)
Membrane Proteins/metabolism , Muscle Development , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Cell Differentiation , Cell Line , Dysferlin , Models, Biological , Muscle Fibers, Skeletal , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Muscular Diseases/pathology , Protein Transport , Sarcolemma/pathology , Wounds and Injuries
16.
PLoS Biol ; 4(8): e242, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16802858

ABSTRACT

Saccharomyces cerevisiae Scc2 binds Scc4 to form an essential complex that loads cohesin onto chromosomes. The prevalence of Scc2 orthologs in eukaryotes emphasizes a conserved role in regulating sister chromatid cohesion, but homologs of Scc4 have not hitherto been identified outside certain fungi. Some metazoan orthologs of Scc2 were initially identified as developmental gene regulators, such as Drosophila Nipped-B, a regulator of cut and Ultrabithorax, and delangin, a protein mutant in Cornelia de Lange syndrome. We show that delangin and Nipped-B bind previously unstudied human and fly orthologs of Caenorhabditis elegans MAU-2, a non-axis-specific guidance factor for migrating cells and axons. PSI-BLAST shows that Scc4 is evolutionarily related to metazoan MAU-2 sequences, with the greatest homology evident in a short N-terminal domain, and protein-protein interaction studies map the site of interaction between delangin and human MAU-2 to the N-terminal regions of both proteins. Short interfering RNA knockdown of human MAU-2 in HeLa cells resulted in precocious sister chromatid separation and in impaired loading of cohesin onto chromatin, indicating that it is functionally related to Scc4, and RNAi analyses show that MAU-2 regulates chromosome segregation in C. elegans embryos. Using antisense morpholino oligonucleotides to knock down Xenopus tropicalis delangin or MAU-2 in early embryos produced similar patterns of retarded growth and developmental defects. Our data show that sister chromatid cohesion in metazoans involves the formation of a complex similar to the Scc2-Scc4 interaction in the budding yeast. The very high degree of sequence conservation between Scc4 homologs in complex metazoans is consistent with increased selection pressure to conserve additional essential functions, such as regulation of cell and axon migration during development.


Subject(s)
Axons/physiology , Cell Movement , Chromatids/physiology , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Conserved Sequence , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/metabolism , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Molecular Sequence Data , Proteins/metabolism , RNA Interference , Saccharomyces cerevisiae , Saccharomyces cerevisiae Proteins/metabolism , Two-Hybrid System Techniques , Xenopus
17.
Eur J Hum Genet ; 13(6): 721-30, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15827562

ABSTRACT

Mutations in dysferlin, a member of the fer1-like protein family that plays a role in membrane integrity and repair, can give rise to a spectrum of neuromuscular disorders with phenotypic variability including limb-girdle muscular dystrophy 2B, Myoshi myopathy and distal anterior compartment myopathy. To improve the tools available for understanding the pathogenesis of the dysferlinopathies, we have established a large source of highly specific antibody reagents against dysferlin by selection of heavy-chain antibody fragments originating from a nonimmune llama-derived phage-display library. By utilizing different truncated forms of recombinant dysferlin for selection and diverse selection methodologies, antibody fragments with specificity for two different dysferlin domains could be identified. The selected llama antibody fragments are functional in Western blotting, immunofluorescence microscopy and immunoprecipitation applications. Using these antibody fragments, we found that calpain 3, which shows a secondary reduction in the dysferlinopathies, interacts with dysferlin.


Subject(s)
Membrane Proteins/analysis , Membrane Proteins/immunology , Muscle Proteins/analysis , Muscle Proteins/immunology , Muscular Dystrophies/metabolism , Animals , Blotting, Western , Calpain/analysis , Camelids, New World/immunology , DNA Mutational Analysis , Dysferlin , Humans , Immunoassay , Immunoglobulin Fragments , Immunoglobulin Heavy Chains , Immunoprecipitation , Isoenzymes/analysis , Microscopy, Fluorescence , Peptide Library , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...