Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Genet Med ; 21(11): 2512-2520, 2019 11.
Article in English | MEDLINE | ID: mdl-31105274

ABSTRACT

PURPOSE: Limb-girdle muscular dystrophies (LGMD) are a genetically heterogeneous category of autosomal inherited muscle diseases. Many genes causing LGMD have been identified, and clinical trials are beginning for treatment of some genetic subtypes. However, even with the gene-level mechanisms known, it is still difficult to get a robust and generalizable prevalence estimation for each subtype due to the limited amount of epidemiology data and the low incidence of LGMDs. METHODS: Taking advantage of recently published exome and genome sequencing data from the general population, we used a Bayesian method to develop a robust disease prevalence estimator. RESULTS: This method was applied to nine recessive LGMD subtypes. The estimated disease prevalence calculated by this method was largely comparable with published estimates from epidemiological studies; however, it highlighted instances of possible underdiagnosis for LGMD2B and 2L. CONCLUSION: The increasing size of aggregated population variant databases will allow for robust and reproducible prevalence estimates of recessive disease, which is critical for the strategic design and prioritization of clinical trials.


Subject(s)
Muscular Dystrophies, Limb-Girdle/epidemiology , Muscular Dystrophies, Limb-Girdle/genetics , Bayes Theorem , Chromosome Mapping , Databases, Genetic , Exome , Female , Humans , Male , Mutation , Prevalence
2.
J Pharmacol Exp Ther ; 364(3): 409-419, 2018 03.
Article in English | MEDLINE | ID: mdl-29284661

ABSTRACT

Ibuprofen, a nonsteroidal anti-inflammatory drug, and nitric oxide (NO) donors have been reported to reduce the severity of muscular dystrophies in mice associated with the absence of dystrophin or α-sarcoglycan, but their effects on mice that are dystrophic due to the absence of dysferlin have not been examined. We have tested ibuprofen, as well as isosorbide dinitrate (ISDN), a NO donor, to learn whether used alone or together they protect dysferlin-null muscle in A/J mice from large strain injury (LSI) induced by a series of high strain lengthening contractions. Mice were maintained on chow containing ibuprofen and ISDN for 4 weeks. They were then subjected to LSI and maintained on the drugs for 3 additional days. We measured loss of torque immediately following injury and at day 3 postinjury, fiber necrosis, and macrophage infiltration at day 3 postinjury, and serum levels of the drugs at the time of euthanasia. Loss of torque immediately after injury was not altered by the drugs. However, the torque on day 3 postinjury significantly decreased as a function of ibuprofen concentration in the serum (range, 0.67-8.2 µg/ml), independent of ISDN. The effects of ISDN on torque loss at day 3 postinjury were not significant. In long-term studies of dysferlinopathic BlAJ mice, lower doses of ibuprofen had no effects on muscle morphology, but reduced treadmill running by 40%. Our results indicate that ibuprofen can have deleterious effects on dysferlin-null muscle and suggest that its use at pharmacological doses should be avoided by individuals with dysferlinopathies.


Subject(s)
Dysferlin/deficiency , Ibuprofen/pharmacology , Muscle, Skeletal/drug effects , Animals , Dysferlin/genetics , Mice , Mice, Knockout , Time Factors
3.
Hum Gene Ther ; 29(7): 749-762, 2018 07.
Article in English | MEDLINE | ID: mdl-28707952

ABSTRACT

Dysferlinopathies comprise a family of disorders caused by mutations in the dysferlin (DYSF) gene, leading to a progressive dystrophy characterized by chronic muscle fiber loss, fat replacement, and fibrosis. To correct the underlying histopathology and function, expression of full-length DYSF is required. Dual adeno-associated virus vectors have been developed, defined by a region of homology, to serve as a substrate for reconstitution of the full 6.5 kb dysferlin cDNA. Previous work studied the efficacy of this treatment through intramuscular and regional delivery routes. To maximize clinical efficacy, dysferlin-deficient mice were treated systemically to target all muscles through the vasculature for efficacy and safety studies. Mice were evaluated at multiple time points between 4 and 13 months post treatment for dysferlin expression and functional improvement using magnetic resonance imaging and magnetic resonance spectroscopy and membrane repair. A systemic dose of 6 × 1012 vector genomes resulted in widespread gene expression in the muscles. Treated muscles showed a significant decrease in central nucleation, collagen deposition, and improvement of membrane repair to wild-type levels. Treated gluteus muscles were significantly improved compared to placebo-treated muscles and were equivalent to wild type in volume, intra- and extramyocellular lipid accumulation, and fat percentage using magnetic resonance imaging and magnetic resonance spectroscopy. Dual-vector treatment allows for production of full-length functional dysferlin with no toxicity. This confirms previous safety data and validates translation of systemic gene delivery for dysferlinopathy patients.


Subject(s)
DNA, Complementary/administration & dosage , Dysferlin/genetics , Genetic Therapy , Muscular Dystrophies, Limb-Girdle/therapy , Animals , DNA, Complementary/genetics , Dependovirus/genetics , Disease Models, Animal , Dysferlin/administration & dosage , Gene Expression Regulation , Genetic Vectors/therapeutic use , Humans , Male , Mice , Muscle, Skeletal , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Mutation
4.
Neuromuscul Disord ; 24(3): 277-87, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24480524

ABSTRACT

The 2013 Dysferlin Conference, sponsored and organized by the Jain Foundation, was held from April 3-6, 2013 in Arlington, VA. Participants included 34 researcher speakers, 5 dysferlinopathy patients and all 8 members of the Jain Foundation team. Dysferlinopathy is a rare disease that typically robs patients of mobility during their second or third decade of life. The goals of these Dysferlin Conferences are to bring experts in the field together so that they will collaborate with one another, to quicken the pace of understanding the biology of the disease and to build effective platforms to ameliorate disease. This is important because the function of dysferlin and how to compensate for its absence is still not well understood, in spite of the fact that the dysferlin gene was identified more than a decade ago. The objective of this conference, therefore, was to share and discuss the newest unpublished research defining the role of dysferlin in skeletal muscle, why its absence causes muscular dystrophy and possible therapies for dysferlin-deficient muscular dystrophy patients.


Subject(s)
Membrane Proteins/physiology , Muscle Proteins/physiology , Muscular Dystrophies, Limb-Girdle , Dysferlin , Humans
5.
PLoS One ; 7(11): e49722, 2012.
Article in English | MEDLINE | ID: mdl-23152929

ABSTRACT

Microtubule-associated proteins of the MAP1 family (MAP1A, MAP1B, and MAP1S) share, among other features, a highly conserved COOH-terminal domain approximately 125 amino acids in length. We conducted a yeast 2-hybrid screen to search for proteins interacting with this domain and identified α1-syntrophin, a member of a multigene family of adapter proteins involved in signal transduction. We further demonstrate that the interaction between the conserved COOH-terminal 125-amino acid domain (which is located in the light chains of MAP1A, MAP1B, and MAP1S) and α1-syntrophin is direct and occurs through the pleckstrin homology domain 2 (PH2) and the postsynaptic density protein 95/disk large/zonula occludens-1 protein homology domain (PDZ) of α1-syntrophin. We confirmed the interaction of MAP1B and α1-syntrophin by co-localization of the two proteins in transfected cells and by co-immunoprecipitation experiments from mouse brain. In addition, we show that MAP1B and α1-syntrophin partially co-localize in Schwann cells of the murine sciatic nerve during postnatal development and in the adult. However, intracellular localization of α1-syntrophin and other Schwann cell proteins such as ezrin and dystrophin-related protein 2 (DRP2) and the localization of the axonal node of Ranvier-associated protein Caspr1/paranodin were not affected in MAP1B null mice. Our findings add to a growing body of evidence that classical MAPs are likely to be involved in signal transduction not only by directly modulating microtubule function, but also through their interaction with signal transduction proteins.


Subject(s)
Calcium-Binding Proteins/metabolism , Central Nervous System/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Muscle Proteins/metabolism , Peripheral Nervous System/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Central Nervous System/cytology , Cytoskeletal Proteins/metabolism , Mice , Microtubules/metabolism , Peripheral Nervous System/cytology , Protein Binding , Protein Transport , Schwann Cells/metabolism
9.
Am J Hum Genet ; 83(6): 714-24, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19026398

ABSTRACT

We have previously reported a group of patients with congenital onset weakness associated with a deficiency of members of the syntrophin-alpha-dystrobrevin subcomplex and have demonstrated that loss of syntrophin and dystrobrevin from the sarcolemma of skeletal muscle can also be associated with denervation. Here, we have further studied four individuals from a consanguineous Egyptian family with a lethal congenital myopathy inherited in an autosomal-recessive fashion and characterized by a secondary loss of beta2-syntrophin and alpha-dystrobrevin from the muscle sarcolemma, central nervous system involvement, and fetal akinesia. We performed homozygosity mapping and candidate gene analysis and identified a mutation that segregates with disease within CNTN1, the gene encoding for the neural immunoglobulin family adhesion molecule, contactin-1. Contactin-1 transcripts were markedly decreased on gene-expression arrays of muscle from affected family members compared to controls. We demonstrate that contactin-1 is expressed at the neuromuscular junction (NMJ) in mice and man in addition to the previously documented expression in the central and peripheral nervous system. In patients with secondary dystroglycanopathies, we show that contactin-1 is abnormally localized to the sarcolemma instead of exclusively at the NMJ. The cntn1 null mouse presents with ataxia, progressive muscle weakness, and postnatal lethality, similar to the affected members in this family. We propose that loss of contactin-1 from the NMJ impairs communication or adhesion between nerve and muscle resulting in the severe myopathic phenotype. This disorder is part of the continuum in the clinical spectrum of congenital myopathies and congenital myasthenic syndromes.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Muscle, Skeletal/pathology , Mutation , Myasthenic Syndromes, Congenital/genetics , Neuromuscular Junction/genetics , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Case-Control Studies , Chromosome Breakage , Chromosome Mapping , Chromosomes, Human, Pair 12 , Cohort Studies , Consanguinity , Conserved Sequence , Contactin 1 , Contactins , DNA Mutational Analysis , Dystrophin-Associated Proteins/genetics , Dystrophin-Associated Proteins/metabolism , Female , Genetic Linkage , Genetic Markers , Haplotypes , Homozygote , Humans , Immunohistochemistry , Infant , Male , Microsatellite Repeats , Molecular Sequence Data , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscle, Skeletal/ultrastructure , Myasthenic Syndromes, Congenital/metabolism , Neuromuscular Junction/metabolism , Pedigree , Sarcolemma/metabolism , Sarcomeres/pathology , Sarcomeres/ultrastructure
10.
Glia ; 56(6): 611-8, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18286648

ABSTRACT

Cytoskeletal scaffolding complexes help organize specialized membrane domains with unique functions on the surface of cells. In this study, we define the scaffolding potential of the Schwann cell dystrophin glycoprotein complex (DGC) by establishing the presence of four syntrophin isoforms, (alpha1, beta1, beta2, and gamma2), and one dystrobrevin isoform, (alpha-dystrobrevin-1), in the abaxonal membrane. Furthermore, we demonstrate the existence of two separate DGCs in Schwann cells that divide the abaxonal membrane into spatially distinct domains, the DRP2/periaxin rich plaques and the Cajal bands that contain Dp116, utrophin, alpha-dystrobrevin-1 and four syntrophin isoforms. Finally, we show that the two different DGCs can scaffold unique accessory molecules in distinct areas of the Schwann cell membrane. Specifically, the cholesterol transporter ABCA1, associates with the Dp116/syntrophin complex in Cajal bands and is excluded from the DRP2/periaxin rich plaques.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Membrane/metabolism , Dystrophin-Associated Proteins , Dystrophin/physiology , Schwann Cells/ultrastructure , ATP Binding Cassette Transporter 1 , Animals , Calcium-Binding Proteins/deficiency , Dystrophin-Associated Proteins/deficiency , Immunoprecipitation/methods , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Molecular Sequence Data , Muscle Proteins/deficiency , Rats , Rats, Wistar , Sciatic Nerve/cytology
11.
J Cell Sci ; 121(Pt 1): 48-54, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18057022

ABSTRACT

alpha-Dystrobrevin associates with and is a homologue of dystrophin, the protein linked to Duchenne and Becker muscular dystrophies. We used a transgenic approach to restore alpha-dystrobrevin to the sarcolemma in mice that lack dystrophin (mdx mice) to study two interrelated functions: (1) the ability of alpha-dystrobrevin to rescue components of the dystrophin complex in the absence of dystrophin and (2) the ability of sarcolemmal alpha-dystrobrevin to ameliorate the dystrophic phenotype. We generated transgenic mice expressing alpha-dystrobrevin-2a linked to a palmitoylation signal sequence and bred them onto the alpha-dystrobrevin-null and mdx backgrounds. Expression of palmitoylated alpha-dystrobrevin prevented the muscular dystrophy observed in the alpha-dystrobrevin-null mice, demonstrating that the altered form of alpha-dystrobrevin was functional. On the mdx background, the palmitoylated form of alpha-dystrobrevin was expressed on the sarcolemma but did not significantly ameliorate the muscular dystrophy phenotype. Palmitoylated dystrobrevin restored alpha-syntrophin and aquaporin-4 (AQP4) to the mdx sarcolemma but was unable to recruit beta-dystroglycan or the sarcoglycans. Despite restoration of sarcolemmal alpha-syntrophin, neuronal nitric oxide synthase (nNOS) was not localized to the sarcolemma, suggesting that nNOS requires both dystrophin and alpha-syntrophin for correct localization. Thus, although nNOS and AQP4 both require interaction with the PDZ domain of alpha-syntrophin for sarcolemmal association, their localization is regulated differentially.


Subject(s)
Aquaporin 4/genetics , Dystrophin-Associated Proteins/genetics , Dystrophin/genetics , Neuropeptides/genetics , Nitric Oxide Synthase Type I/genetics , Sarcolemma/metabolism , Animals , Dystrophin/chemistry , Dystrophin/metabolism , Dystrophin-Associated Proteins/metabolism , Lipoylation , Mice , Mice, Inbred mdx , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Muscles/metabolism , Muscles/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Neuropeptides/metabolism , Nitric Oxide Synthase Type I/metabolism , PDZ Domains , Protein Binding , Sarcolemma/chemistry
12.
Gene Expr ; 14(1): 47-57, 2007.
Article in English | MEDLINE | ID: mdl-17933218

ABSTRACT

The dystrophin glycoprotein complex (DGC) is critical for muscle stability, and mutations in DGC proteins lead to muscular dystrophy. The DGC also contributes to the maturation and maintenance of the neuromuscular junction (NMJ). The gene encoding the DGC protein alpha-dystrobrevin undergoes alternative splicing to produce at least five known isoforms. Isoform-specific antibody staining and reverse transcription PCR in mutant mice with a deletion of exon 3 of the alpha-dystrobrevin gene suggested the existence of a remaining synaptic isoform, which might be compensating for alpha-dystrobrevin function. To test this possibility and to more completely understand the synaptic function of alpha-dystrobrevin, we used a two-step homologous recombination strategy combined with in vivo Cre-mediated excision to generate mice with a large deletion of the alpha-dystrobrevin gene to disrupt all isoforms. However, these mice did not exhibit a more severe NMJ phenotype than that observed in the exon 3-deleted mice. Nonetheless, these mice not only eliminate possible compensation by remaining isoforms of alpha-dystrobrevin, but also offer a conditional allele that could be used to identify tissue-specific and developmental functions of alpha-dystrobrevin. This work also demonstrates a successful strategy to achieve deletion of a large genomic sequence, which can be a valuable tool for functional studies of genes encoding multiple isoforms that span a large genomic region.


Subject(s)
Dystrophin-Associated Proteins/genetics , Gene Deletion , Neuromuscular Junction/metabolism , Neuropeptides/genetics , Animals , Blotting, Northern , Exons , Immunohistochemistry , Mice , Mice, Mutant Strains , Phenotype , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Exp Cell Res ; 312(16): 3084-95, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16857187

ABSTRACT

The syntrophins are a family of scaffolding proteins with multiple protein interaction domains that link signaling proteins to dystrophin family members. Each of the three most characterized syntrophins (alpha, beta1, beta2) contains a PDZ domain that binds a unique set of signaling proteins including kinases, ion and water channels, and neuronal nitric oxide synthase (nNOS). The PDZ domains of the gamma-syntrophins do not bind nNOS. In vitro pull-down assays show that the gamma-syntrophins can bind dystrophin but have unique preferences for the syntrophin binding sites of dystrophin family members. Despite their ability to bind dystrophin in vitro, neither gamma-syntrophin isoform co-localizes with dystrophin in skeletal muscle. Furthermore, gamma-syntrophins do not co-purify with dystrophin isolated from mouse tissue. These data suggest that the interaction of gamma-syntrophin with dystrophin is transient and potentially subject to regulatory mechanisms. gamma1-Syntrophin is highly expressed in brain and is specifically localized in hippocampal pyramidal neurons, Purkinje neurons in cerebellum, and cortical neurons. gamma2-Syntrophin is expressed in many tissues including skeletal muscle where it is found only in the subsynaptic space beneath the neuromuscular junction. In both neurons and muscle, gamma-syntrophin isoforms localize to the endoplasmic reticulum where they may form a scaffold for signaling and trafficking.


Subject(s)
Dystrophin-Associated Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies/immunology , Dystrophin/metabolism , Dystrophin-Associated Proteins/chemistry , Gene Expression Profiling , Humans , Mice , Molecular Sequence Data , Neurons/cytology , Nitric Oxide Synthase Type I/metabolism , Protein Binding , Protein Isoforms/metabolism , Protein Structure, Tertiary , Protein Transport , Purkinje Cells/cytology , Sarcoplasmic Reticulum/metabolism , Sequence Homology, Amino Acid
14.
J Biol Chem ; 279(8): 7014-23, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14623885

ABSTRACT

Mice rendered null for alpha-dystrobrevin, a component of the dystrophin complex, have muscular dystrophy, despite the fact that the sarcolemma remains relatively intact (Grady, R. M., Grange, R. W., Lau, K. S., Maimone, M. M., Nichol, M. C., Stull, J. T., and Sanes, J. R. (1999) Nat. Cell Biol. 1, 215-220) Thus, alpha-dystrobrevin may serve a signaling function that is important for the maintenance of muscle integrity. We have identified a new dystrobrevin-associated protein, DAMAGE, that may play a signaling role in brain, muscle, and peripheral nerve. In humans, DAMAGE is encoded by an intronless gene located at chromosome Xq13.1, a locus that contains genes involved in mental retardation. DAMAGE associates directly with alpha-dystrobrevin, as shown by yeast two-hybrid, and co-immunoprecipitates with the dystrobrevin-syntrophin complex from brain. This co-immunoprecipitation is dependent on the presence of alpha-dystrobrevin but not beta-dystrobrevin. The DAMAGE protein contains a potential nuclear localization signal, 30 12-amino acid repeats, and two MAGE homology domains. The domain structure of DAMAGE is similar to that of NRAGE, a MAGE protein that mediates p75 neurotrophin receptor signaling and neuronal apoptosis (Salehi, A. H., Roux, P. P., Kubu, C. J., Zeindler, C., Bhakar, A., Tannis, L. L., Verdi, J. M., and Barker, P. A. (2000) Neuron 27, 279-288). DAMAGE is highly expressed in brain and is present in the cell bodies and dendrites of hippocampal and Purkinje neurons. In skeletal muscle, DAMAGE is at the postsynaptic membrane and is associated with a subset of myonuclei. DAMAGE is also expressed in peripheral nerve, where it localizes along with other members of the dystrophin complex to the perineurium and myelin. These results expand the role of dystrobrevin and the dystrophin complex in membrane signaling and disease.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/physiology , Cytoskeletal Proteins/chemistry , Dystrophin-Associated Proteins , Dystrophin/chemistry , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/physiology , Amino Acid Sequence , Animals , Apoptosis , Blotting, Northern , Blotting, Western , Brain/metabolism , Carrier Proteins/genetics , Cloning, Molecular , Cytoskeletal Proteins/genetics , DNA/chemistry , DNA, Complementary/metabolism , Humans , Immunohistochemistry , Introns , Macaca , Membrane Proteins/genetics , Mice , Microscopy, Confocal , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscles/metabolism , Myelin Sheath/chemistry , Nerve Tissue Proteins/genetics , Neurons/metabolism , Nuclear Localization Signals , Peripheral Nervous System/metabolism , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Tissue Distribution , Transfection , Two-Hybrid System Techniques , X Chromosome
15.
Neurosignals ; 11(3): 123-9, 2002.
Article in English | MEDLINE | ID: mdl-12138249

ABSTRACT

Dystrophin and its associated proteins were originally identified in skeletal muscle, where the complex provides mechanical stabilization to the sarcolemma during contraction. However, the dystrophin complex is also present at membrane specializations in many non-muscle cells, including synaptic sites in neurons. The function of the dystrophin complex at these sites is still unknown, but emerging results suggest that the dystrophin complex can function as a scaffold for signaling proteins. In this review, we examine the growing body of evidence that suggests the dystrophin complex may have a dual function: membrane stabilization and transmembrane signaling. We focus on the role of two dystrophin-associated proteins, syntrophin and dystrobrevin, in the formation of a signaling scaffold and review evidence suggesting a role in synapse formation and maintenance.


Subject(s)
Cytoskeleton/ultrastructure , Dystrophin-Associated Proteins , Dystrophin/physiology , Membrane Proteins/physiology , Muscle Proteins/physiology , Nerve Tissue Proteins/physiology , Neuropeptides/physiology , Synapses/ultrastructure , Animals , Cell Membrane/physiology , Cell Membrane/ultrastructure , Cytoskeleton/chemistry , Humans , Intellectual Disability/genetics , MAP Kinase Signaling System , Macromolecular Substances , Male , Membrane Proteins/chemistry , Muscle Proteins/chemistry , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/psychology , Neuropeptides/chemistry , Neuropeptides/deficiency , Neuropeptides/genetics , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Organ Specificity , Protein Structure, Tertiary , Synapses/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...