Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
Nat Commun ; 15(1): 4935, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858388

ABSTRACT

Cell polarity mechanisms allow the formation of specialized membrane domains with unique protein compositions, signalling properties, and functional characteristics. By analyzing the localization of potassium channels and proteins belonging to the dystrophin-associated protein complex, we reveal the existence of distinct planar-polarized membrane compartments at the surface of C. elegans muscle cells. We find that muscle polarity is controlled by a non-canonical Wnt signalling cascade involving the ligand EGL-20/Wnt, the receptor CAM-1/Ror, and the intracellular effector DSH-1/Dishevelled. Interestingly, classical planar cell polarity proteins are not required for this process. Using time-resolved protein degradation, we demonstrate that -while it is essentially in place by the end of embryogenesis- muscle polarity is a dynamic state, requiring continued presence of DSH-1 throughout post-embryonic life. Our results reveal the unsuspected complexity of the C. elegans muscle membrane and establish a genetically tractable model system to study cellular polarity and membrane compartmentalization in vivo.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cell Polarity , Dystrophin , Muscles , Wnt Signaling Pathway , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Dystrophin/metabolism , Dystrophin/genetics , Muscles/metabolism , Dishevelled Proteins/metabolism , Dishevelled Proteins/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Cell Membrane/metabolism , Dystrophin-Associated Protein Complex/metabolism , Dystrophin-Associated Protein Complex/genetics , Wnt Proteins/metabolism , Signal Transduction
2.
Mass Spectrom Rev ; 43(1): 90-105, 2024.
Article in English | MEDLINE | ID: mdl-36420714

ABSTRACT

The dystrophin-associated protein complex (DAPC) is a highly organized multiprotein complex that plays a pivotal role in muscle fiber structure integrity and cell signaling. The complex is composed of three distinct interacting subgroups, intracellular peripheral proteins, transmembrane glycoproteins, and extracellular glycoproteins subcomplexes. Dystrophin protein nucleates the DAPC and is important for connecting the intracellular actin cytoskeletal filaments to the sarcolemma glycoprotein complex that is connected to the extracellular matrix via laminin, thus stabilizing the sarcolemma during muscle fiber contraction and relaxation. Genetic mutations that lead to lack of expression or altered expression of any of the DAPC proteins are associated with different types of muscle diseases. Hence characterization of this complex in healthy and dystrophic muscle might bring insights into its role in muscle pathogenesis. This review highlights the role of mass spectrometry in characterizing the DAPC interactome as well as post-translational glycan modifications of some of its components such as α-dystroglycan. Detection and quantification of dystrophin using targeted mass spectrometry are also discussed in the context of healthy versus dystrophic skeletal muscle.


Subject(s)
Dystrophin-Associated Protein Complex , Dystrophin , Dystrophin/analysis , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/analysis , Dystrophin-Associated Protein Complex/metabolism , Laminin/analysis , Laminin/metabolism , Sarcolemma/chemistry , Sarcolemma/metabolism , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Glycoproteins/analysis
3.
Gene Ther ; 29(9): 520-535, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35105949

ABSTRACT

Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the gene encoding dystrophin. Gene therapy using micro-dystrophin (MD) transgenes and recombinant adeno-associated virus (rAAV) vectors hold great promise. To overcome the limited packaging capacity of rAAV vectors, most MD do not include dystrophin carboxy-terminal (CT) domain. Yet, the CT domain is known to recruit α1- and ß1-syntrophins and α-dystrobrevin, a part of the dystrophin-associated protein complex (DAPC), which is a signaling and structural mediator of muscle cells. In this study, we explored the impact of inclusion of the dystrophin CT domain on ΔR4-23/ΔCT MD (MD1), in DMDmdx rats, which allows for relevant evaluations at muscular and cardiac levels. We showed by LC-MS/MS that MD1 expression is sufficient to restore the interactions at a physiological level of most DAPC partners in skeletal and cardiac muscles, and that inclusion of the CT domain increases the recruitment of some DAPC partners at supra-physiological levels. In parallel, we demonstrated that inclusion of the CT domain does not improve MD1 therapeutic efficacy on DMD muscle and cardiac pathologies. Our work highlights new evidences of the therapeutic potential of MD1 and strengthens the relevance of this candidate for gene therapy of DMD.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Animals , Chromatography, Liquid , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/metabolism , Genetic Therapy , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Rats , Tandem Mass Spectrometry
4.
Protein Expr Purif ; 167: 105525, 2020 03.
Article in English | MEDLINE | ID: mdl-31682967

ABSTRACT

Mutation of the gene encoding γ-sarcoglycan (SGCG), an integral membrane protein responsible for maintaining the integrity of the muscle cell sarcolemma, results in Limb-Girdle Muscular Dystrophy (LGMD), a congenital disease with no current treatment options. This member of the sarcoglycan glycoprotein family is a vital component of the Dystrophin Complex, which together facilitate normal muscle function. However, very little is known about the structure and dynamics of these proteins, and of membrane glycoproteins in general. This is due to a number of factors, including their complexity, heterogeneity and highly-specific native environments. The expression, purification, and structural study of membrane proteins is further impeded by their hydrophobic nature and consequent propensity to aggregate in aqueous solutions. Here, we report the first successful expression and purification of milligram quantities of full-length recombinant SGCG, utilizing fusion protein-guided overexpression to inclusion bodies in Escherichia coli. Purification of SGCG from the fusion protein, TrpΔLE, was facilitated using chemical cleavage. Cleavage products were then isolated by size-exclusion chromatography. Successful purification of the protein was confirmed using SDS-PAGE and mass spectroscopy. Finally, solution nuclear magnetic resonance spectroscopy of uniformly 15N-labeled SGCG in detergent environments was performed, yielding the first spectra of the full-length membrane glycoprotein, SGCG. These results represent the initial structural studies of SGCG, laying the foundation for further investigation on the interaction and dynamics of other integral membrane proteins. More specifically, this data allows for opportunities in the future for enhanced treatment modalities and cures for LGMD.


Subject(s)
Sarcoglycans , Chromatography, Gel , Cloning, Molecular/methods , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/isolation & purification , Dystrophin-Associated Protein Complex/metabolism , Escherichia coli , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Glycoproteins/genetics , Glycoproteins/isolation & purification , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Membrane Proteins/analysis , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/etiology , Mutation , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Sarcoglycans/biosynthesis , Sarcoglycans/chemistry , Sarcoglycans/genetics , Sarcoglycans/isolation & purification , Sarcolemma/metabolism , Solubility
5.
Sci Rep ; 9(1): 2770, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808964

ABSTRACT

The dystrophin-glycoprotein complex (DGC) links the muscle cytoskeleton to the extracellular matrix and is responsible for force transduction and protects the muscle fibres from contraction induced damage. Mutations in components of the DGC are responsible for muscular dystrophies and congenital myopathies. Expression of DGC components have been shown to be altered in many myopathies. In contrast we have very little evidence of whether adaptive changes in muscle impact on DGC expression. In this study we investigated connection between muscle fibre phenotype and the DGC. Our study reveals that the levels of DGC proteins at the sarcolemma differ in highly glycolytic muscle compared to wild-type and that these changes can be normalised by the super-imposition of an oxidative metabolic programme. Importantly we show that the metabolic properties of the muscle do not impact on the total amount of DGC components at the protein level. Our work shows that the metabolic property of a muscle fibre is a key factor in regulating the expression of DGC proteins at the sarcolemma.


Subject(s)
Dystrophin-Associated Protein Complex/metabolism , Dystrophin/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Collagen Type IV/metabolism , Laminin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Myostatin/deficiency , Myostatin/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Sarcoglycans/metabolism
6.
Methods Mol Biol ; 1828: 327-342, 2018.
Article in English | MEDLINE | ID: mdl-30171551

ABSTRACT

Exon-skipping antisense oligonucleotides (AOs) are promising treatments for muscle-related genetic ailments including Duchenne muscular dystrophy (DMD), but clinical translation is unfortunately hampered by insufficient systemic delivery. Here we describe that how one can employ a glucose-fructose injection mixture to improve muscle uptake and functional outcomes of DMD AOs in energy-deficient peripheral muscles of mdx mice. The potentiating effect of glucose-fructose on AOs in energy-deficient muscles offers a simple and economical method for enhancing AO potency, reducing screening costs for researchers and accelerating the translation of nucleic acid-based therapeutics in DMD and other muscular dystrophies.


Subject(s)
Exons , Fructose/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , RNA Splicing , Adenosine Triphosphate/metabolism , Animals , Clathrin/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/genetics , Dystrophin-Associated Protein Complex/metabolism , Endocytosis , Energy Metabolism , Fructose/metabolism , Glucose/metabolism , Humans , Immunohistochemistry , Metabolic Networks and Pathways , Mice , Mice, Inbred mdx , Morpholinos/administration & dosage , Morpholinos/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics
7.
Biomaterials ; 183: 54-66, 2018 11.
Article in English | MEDLINE | ID: mdl-30149230

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is a fatal genetic disorder currently having no cure. Here we report that culture substrates patterned with nanogrooves and functionalized with Matrigel (or laminin) present an engineered cell microenvironment to allow myotubes derived from non-diseased, less-affected DMD, and severely-affected DMD human induced pluripotent stem cells (hiPSCs) to exhibit prominent differences in alignment and orientation, providing a sensitive phenotypic biomarker to potentially facilitate DMD drug development and early diagnosis. We discovered that myotubes differentiated from myogenic progenitors derived from non-diseased hiPSCs align nearly perpendicular to nanogrooves, a phenomenon not reported previously. We further found that myotubes derived from hiPSCs of a dystrophin-null DMD patient orient randomly, and those from hiPSCs of a patient carrying partially functional dystrophin align approximately 14° off the alignment direction of non-diseased myotubes. Substrates engineered with micron-scale grooves and/or cell adhesion molecules only interacting with integrins all guide parallel myotube alignment to grooves and lose the ability to distinguish different cell types. Disruption of the interaction between the Dystrophin-Associated-Protein-Complex (DAPC) and laminin by heparin or anti-α-dystroglycan antibody IIH6 disenables myotubes to align perpendicular to nanogrooves, suggesting that this phenotype is controlled by the DAPC-mediated cytoskeleton-extracellular matrix linkage.


Subject(s)
Muscle Fibers, Skeletal/physiology , Muscular Dystrophy, Duchenne/diagnosis , Nanostructures/chemistry , Biomarkers , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Collagen/metabolism , Cytoskeleton/metabolism , Drug Combinations , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/metabolism , Extracellular Matrix/metabolism , Humans , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/physiology , Laminin/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myoblasts/metabolism , Myoblasts/pathology , Phenotype , Proteoglycans/metabolism
8.
Matrix Biol ; 68-69: 628-636, 2018 08.
Article in English | MEDLINE | ID: mdl-29475025

ABSTRACT

Endplate acetylcholinesterase (AChE) deficiency is a form of congenital myasthenic syndrome (CMS) caused by mutations in COLQ, which encodes collagen Q (ColQ). ColQ is an extracellular matrix (ECM) protein that anchors AChE to the synaptic basal lamina. Biglycan, encoded by BGN, is another ECM protein that binds to the dystrophin-associated protein complex (DAPC) on skeletal muscle, which links the actin cytoskeleton and ECM proteins to stabilize the sarcolemma during repeated muscle contractions. Upregulation of biglycan stabilizes the DPAC. Gene therapy can potentially ameliorate any disease that can be recapitulated in cultured cells. However, the difficulty of tissue-specific and developmental stage-specific regulated expression of transgenes, as well as the difficulty of introducing a transgene into all cells in a specific tissue, prevents us from successfully applying gene therapy to many human diseases. In contrast to intracellular proteins, an ECM protein is anchored to the target tissue via its specific binding affinity for protein(s) expressed on the cell surface within the target tissue. Exploiting this unique feature of ECM proteins, we developed protein-anchoring therapy in which a transgene product expressed even in remote tissues can be delivered and anchored to a target tissue using specific binding signals. We demonstrate the application of protein-anchoring therapy to two disease models. First, intravenous administration of adeno-associated virus (AAV) serotype 8-COLQ to Colq-deficient mice, resulting in specific anchoring of ectopically expressed ColQ-AChE at the NMJ, markedly improved motor functions, synaptic transmission, and the ultrastructure of the neuromuscular junction (NMJ). In the second example, Mdx mice, a model for Duchenne muscular dystrophy, were intravenously injected with AAV8-BGN. The treatment ameliorated motor deficits, mitigated muscle histopathologies, decreased plasma creatine kinase activities, and upregulated expression of utrophin and DAPC component proteins. We propose that protein-anchoring therapy could be applied to hereditary/acquired defects in ECM and secreted proteins, as well as therapeutic overexpression of such factors.


Subject(s)
Acetylcholinesterase/genetics , Biglycan/genetics , Collagen/genetics , Genetic Therapy/methods , Muscle Proteins/genetics , Muscular Dystrophy, Duchenne/therapy , Myasthenic Syndromes, Congenital/therapy , Acetylcholinesterase/metabolism , Actin Cytoskeleton/metabolism , Animals , Biglycan/metabolism , Collagen/metabolism , Dependovirus/chemistry , Disease Models, Animal , Dystrophin-Associated Protein Complex/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , GPI-Linked Proteins/metabolism , Genetic Vectors/administration & dosage , Humans , Mice , Muscle Proteins/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Myasthenic Syndromes, Congenital/genetics , Myasthenic Syndromes, Congenital/metabolism
9.
J Cell Physiol ; 233(7): 5142-5159, 2018 07.
Article in English | MEDLINE | ID: mdl-28464259

ABSTRACT

Dystrophin protein in association with several other cellular proteins and glycoproteins leads to the formation of a large multifaceted protein complex at the cell membrane referred to as dystrophin glycoprotein complex (DGC), that serves distinct functions in cell signaling and maintaining the membrane stability as well as integrity. In accordance with this, several findings suggest exquisite role of DGC in signaling pathways associated with cell development and/or maintenance of homeostasis. In the present review, we summarize the established facts about the various components of this complex with emphasis on recent insights into specific contribution of the DGC in cell signaling at the membrane. We have also discussed the recent advances made in exploring the molecular associations of DGC components within the cells and the functional implications of these interactions. Our review would help to comprehend the composition, role, and functioning of DGC and may lead to a deeper understanding of its role in several human diseases.


Subject(s)
Cell Membrane/genetics , Dystrophin-Associated Protein Complex/genetics , Dystrophin/genetics , Glycoproteins/genetics , Cell Membrane/chemistry , Dystrophin/chemistry , Dystrophin-Associated Protein Complex/chemistry , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Signal Transduction
10.
J Neurosci Res ; 96(2): 180-193, 2018 02.
Article in English | MEDLINE | ID: mdl-28509351

ABSTRACT

Astrocytes play a critical role in regulating the interface between the cerebral vasculature and the central nervous system. Contributing to this is the astrocytic endfoot domain, a specialized structure that ensheathes the entirety of the vasculature and mediates signaling between endothelial cells, pericytes, and neurons. The astrocytic endfoot has been implicated as a critical element of the glymphatic pathway, and changes in protein expression profiles in this cellular domain are linked to Alzheimer's disease pathology. Despite this, basic physiological properties of this structure remain poorly understood including the developmental timing of its formation, and the protein components that localize there to mediate its functions. Here we use human transcriptome data from male and female subjects across several developmental stages and brain regions to characterize the gene expression profile of the dystrophin-associated complex (DAC), a known structural component of the astrocytic endfoot that supports perivascular localization of the astroglial water channel aquaporin-4. Transcriptomic profiling is also used to define genes exhibiting parallel expression profiles to DAC elements, generating a pool of candidate genes that encode gene products that may contribute to the physiological function of the perivascular astrocytic endfoot domain. We found that several genes encoding transporter proteins are transcriptionally associated with DAC genes.


Subject(s)
Astrocytes/metabolism , Brain/cytology , Brain/growth & development , Dystrophin-Associated Protein Complex/metabolism , Transcriptome/physiology , Adolescent , Adult , Analysis of Variance , Aquaporin 4/metabolism , Calcium-Binding Proteins/metabolism , Child , Dystrophin-Associated Proteins/metabolism , Female , Gene Ontology , Humans , Male , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Neuropeptides/metabolism , Subcellular Fractions/metabolism , Young Adult
11.
Neurosci Lett ; 658: 142-149, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-28855126

ABSTRACT

INTRODUCTION: Multiple components of the dystrophin-associated protein complex (DAPC) are expressed in numerous tissues including the brain. Members of the DAPC and dysbindin are abnormally expressed in the brain of Duchenne Muscular Dystrophy (DMD) patients, which has been associated with cognitive impairments. However, little is known about the expression pattern of individual members of the DAPC in animal models of DMD and their relationship with dysbindin. METHODS: Ten mdx mice were randomly allocated into a control and intervention group [(-)-epicatechin (Epi) 1mg/kg/day for four weeks] and results compared to a wild-type mice. After sacrifice, brain pre-frontal cortices were collected for Western blotting and immunoprecipitation assays, and sagittal sections processed for immunohistochemistry. RESULTS: Epi promotes a partial recovery of DAPC members [α1-Syntrophin, sarcoglycans (SG), dystrophin 71 (Dp71)], dysbindin, and utrophin protein levels. Epi also appears to restore the association of DAPC between dysbindin, and utrophin with Dp71 and ε-SG. Co-immunostaining evidence increased protein levels of dysbindin, dystrophin, and ε-SG and their colocalization. CONCLUSIONS: Altogether, results suggest that Epi is capable of restoring pre-frontal cortex DAPC and dysbindin levels of mdx mice towards that of healthy brains. The functional implications of such studies warrant further investigation.


Subject(s)
Catechin/genetics , Dysbindin/metabolism , Dystrophin-Associated Protein Complex/metabolism , Frontal Lobe/metabolism , Muscular Dystrophy, Duchenne/metabolism , Animals , Dystrophin/metabolism , Mice, Inbred mdx , Utrophin/metabolism
12.
J Vasc Res ; 54(4): 235-245, 2017.
Article in English | MEDLINE | ID: mdl-28768281

ABSTRACT

BACKGROUND: Smooth muscle cells are important for atherosclerotic plaque stability. Their proper ability to communicate with the extracellular matrix is crucial for maintaining the correct tissue integrity. In this study, we have investigated the role of ß-sarcoglycan within the matrix-binding dystrophin-glycoprotein complex in the development of atherosclerosis. RESULTS: Atherosclerotic plaque development was significantly reduced in ApoE-deficient mice lacking ß-sarcoglycan, and their plaques contained an increase in differentiated smooth muscle cells. ApoE-deficient mice lacking ß-sarcoglycan showed a reduction in ovarian adipose tissue and adipocyte size, while the total weight of the animals was not significantly different. Western blot analysis of adipose tissues showed a decreased activation of protein kinase B, while that of AMP-activated kinase was increased in mice lacking ß-sarcoglycan. Analysis of plasma in ß-sarcoglycan-deficient mice revealed reduced levels of leptin, adiponectin, insulin, cholesterol, and triglycerides but increased levels of IL-6, IL-17, and TNF-α. CONCLUSIONS: Our results indicate that the dystrophin-glycoprotein complex and ß-sarcoglycan can affect the atherosclerotic process. Furthermore, the results show the effects of ß-sarcoglycan deficiency on adipose tissue and lipid metabolism, which may also have contributed to the atherosclerotic plaque reduction.


Subject(s)
Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Atherosclerosis/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic , Sarcoglycans/deficiency , AMP-Activated Protein Kinases/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipokines/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Dystrophin-Associated Protein Complex/metabolism , Female , Genetic Predisposition to Disease , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Phenotype , Proto-Oncogene Proteins c-akt/metabolism , Sarcoglycans/genetics
13.
Invest Ophthalmol Vis Sci ; 58(9): 3795-3803, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28744553

ABSTRACT

Purpose: Mutations in genes encoding the dystrophin-associated glycoprotein complex (DGC) can cause muscular dystrophy and disturb synaptic transmission in the photoreceptor ribbon synapse. However, the molecular composition and specific functions of the photoreceptor DGC remain unknown. Brefeldin A-resistant Arf-GEF 2 (BRAG2), also known as IQSEC1, is a guanine nucleotide exchange factor for ADP-ribosylation factor 6 (Arf6), a critical GTPase that regulates endosomal trafficking and actin cytoskeleton remodeling. In the present study, we characterized the expression of BRAG2a, an alternative splicing isoform of BRAG2, in the adult mouse photoreceptor. Methods: Immunofluorescence and immunoelectron microscopic analyses of adult mouse retinas were performed using a novel anti-BRAG2a antibody. Pull-down, immunoprecipitation, and in situ proximity ligation assays were performed to examine the interaction between BRAG2a and the DGC in vivo. Results: Immunofluorescence demonstrated punctate colocalization of BRAG2a with ß-dystroglycan in the outer plexiform layer. Immunoelectron microscopy revealed the localization of BRAG2a at the plasma membrane of lateral walls and processes of photoreceptor terminals within the synaptic cavity. Pull-down and immunoprecipitation assays using retinal lysates demonstrated the protein complex formation between BRAG2a with the DGC. In situ proximity ligation assays further detected a close spatial relationship between BRAG2a and ß-dystroglycan in the outer plexiform layer. Conclusions: The present study provided evidence that BRAG2a is a novel component of the photoreceptor DGC, suggesting functional involvement of the BRAG2a-Arf6 pathway downstream of the DGC.


Subject(s)
ADP-Ribosylation Factors/metabolism , Dystroglycans/metabolism , Dystrophin-Associated Protein Complex/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Presynaptic Terminals/metabolism , ADP-Ribosylation Factor 6 , Alternative Splicing , Animals , Fluorescent Antibody Technique, Indirect , Immunoblotting , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Immunoelectron , Plasmids , Polymerase Chain Reaction , Protein Isoforms
14.
Curr Opin Pharmacol ; 34: 36-48, 2017 06.
Article in English | MEDLINE | ID: mdl-28486179

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of dystrophin, essential for muscle fibre integrity. Despite extensive pre-clinical studies, development of an effective treatment has proved challenging. More recently, significant progress has been made with the first drug approval using a genetic approach and the application of pharmacological agents which slow the progression of the disease. Drug development for DMD has mainly used two strategies: (1) the restoration of dystrophin expression or the expression of the compensatory utrophin protein as an efficient surrogate, and (2) the mitigation of secondary downstream pathological mechanisms. This review details current most promising pharmacological approaches and clinical trials aiming to tackle the pathogenesis of this multifaceted disorder.


Subject(s)
Muscular Dystrophy, Duchenne/drug therapy , Animals , Dystrophin/deficiency , Dystrophin-Associated Protein Complex/metabolism , Humans , Muscular Dystrophy, Duchenne/metabolism
15.
Sci Rep ; 6: 28097, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27323895

ABSTRACT

The development of medical approaches requires preclinical and clinical trials for assessment of therapeutic efficacy. Such evaluation entails the use of biomarkers, which provide information on the response to the therapeutic intervention. One newly-proposed class of biomarkers is the microRNA (miRNA) molecules. In muscular dystrophies (MD), the dysregulation of miRNAs was initially observed in muscle biopsy and later extended to plasma samples, suggesting that they may be of interest as biomarkers. First, we demonstrated that dystromiRs dysregulation occurs in MD with either preserved or disrupted expression of the dystrophin-associated glycoprotein complex, supporting the utilization of dystromiRs as generic biomarkers in MD. Then, we aimed at evaluation of the capacity of miRNAs as monitoring biomarkers for experimental therapeutic approach in MD. To this end, we took advantage of our previously characterized gene therapy approach in a mouse model for α-sarcoglycanopathy. We identified a dose-response correlation between the expression of miRNAs on both muscle tissue and blood serum and the therapeutic benefit as evaluated by a set of new and classically-used evaluation methods. This study supports the utility of profiling circulating miRNAs for the evaluation of therapeutic outcome in medical approaches for MD.


Subject(s)
Biomarkers/blood , Circulating MicroRNA/blood , Muscular Dystrophies/blood , Muscular Dystrophies/diagnosis , Animals , CD11b Antigen/metabolism , Disease Models, Animal , Dystrophin-Associated Protein Complex/genetics , Dystrophin-Associated Protein Complex/metabolism , Genetic Therapy , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Sarcoglycans/genetics
16.
J Clin Invest ; 125(11): 4186-95, 2015 Nov 02.
Article in English | MEDLINE | ID: mdl-26457733

ABSTRACT

Exon skipping uses antisense oligonucleotides as a treatment for genetic diseases. The antisense oligonucleotides used for exon skipping are designed to bypass premature stop codons in the target RNA and restore reading frame disruption. Exon skipping is currently being tested in humans with dystrophin gene mutations who have Duchenne muscular dystrophy. For Duchenne muscular dystrophy, the rationale for exon skipping derived from observations in patients with naturally occurring dystrophin gene mutations that generated internally deleted but partially functional dystrophin proteins. We have now expanded the potential for exon skipping by testing whether an internal, in-frame truncation of a transmembrane protein γ-sarcoglycan is functional. We generated an internally truncated γ-sarcoglycan protein that we have termed Mini-Gamma by deleting a large portion of the extracellular domain. Mini-Gamma provided functional and pathological benefits to correct the loss of γ-sarcoglycan in a Drosophila model, in heterologous cell expression studies, and in transgenic mice lacking γ-sarcoglycan. We generated a cellular model of human muscle disease and showed that multiple exon skipping could be induced in RNA that encodes a mutant human γ-sarcoglycan. Since Mini-Gamma represents removal of 4 of the 7 coding exons in γ-sarcoglycan, this approach provides a viable strategy to treat the majority of patients with γ-sarcoglycan gene mutations.


Subject(s)
Dystrophin-Associated Protein Complex/chemistry , Genetic Therapy , Muscular Dystrophies, Limb-Girdle/therapy , Oligonucleotides, Antisense/therapeutic use , Protein Engineering , Sarcoglycans/genetics , Animals , Codon, Nonsense/genetics , Diaphragm/metabolism , Diaphragm/pathology , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Exons , Fibrosis , HEK293 Cells , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/therapy , Mutation , Myocardium/metabolism , Myocardium/pathology , Oligonucleotides, Antisense/pharmacology , Protein Interaction Mapping , Protein Structure, Tertiary , RNA, Messenger/chemistry , RNA, Messenger/genetics , Recombinant Fusion Proteins/metabolism , Sarcoglycans/biosynthesis , Sarcoglycans/chemistry , Sarcoglycans/deficiency , Sarcolemma/metabolism , Sequence Deletion
17.
PLoS One ; 10(9): e0137328, 2015.
Article in English | MEDLINE | ID: mdl-26378780

ABSTRACT

The precise functional role of the dystrophin 71 in neurons is still elusive. Previously, we reported that dystrophin 71d and dystrophin 71f are present in nuclei from cultured neurons. In the present work, we performed a detailed analysis of the intranuclear distribution of dystrophin 71 isoforms (Dp71d and Dp71f), during the temporal course of 7-day postnatal rats hippocampal neurons culture for 1h, 2, 4, 10, 15 and 21 days in vitro (DIV). By immunofluorescence assays, we detected the highest level of nuclear expression of both dystrophin Dp71 isoforms at 10 DIV, during the temporal course of primary culture. Dp71d and Dp71f were detected mainly in bipolar GABAergic (≥60%) and multipolar Glutamatergic (≤40%) neurons, respectively. We also characterized the existence of two nuclear dystrophin-associated protein complexes (DAPC): dystrophin 71d or dystrophin 71f bound to ß-dystroglycan, α1-, ß-, α2-dystrobrevins, α-syntrophin, and syntrophin-associated protein nNOS (Dp71d-DAPC or Dp71f-DAPC, respectively), in the hippocampal neurons. Furthermore, both complexes were localized in interchromatin granule cluster structures (nuclear speckles) of neuronal nucleoskeleton preparations. The present study evinces that each Dp71's complexes differ slightly in dystrobrevins composition. The results demonstrated that Dp71d-DAPC was mainly localized in bipolar GABAergic and Dp71f-DAPC in multipolar Glutamatergic hippocampal neurons. Taken together, our results show that dystrophin 71d, dystrophin 71f and DAP integrate protein complexes, and both complexes were associated to nuclear speckles structures.


Subject(s)
Cell Nucleus/metabolism , Dystrophin-Associated Protein Complex/genetics , Dystrophin/genetics , GABAergic Neurons/cytology , Hippocampus/cytology , Animals , Calcium-Binding Proteins/metabolism , Cells, Cultured , Dystroglycans/metabolism , Dystrophin-Associated Protein Complex/metabolism , Female , Fluorescent Antibody Technique , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Protein Isoforms/genetics , Rats , Rats, Wistar
18.
Brain Struct Funct ; 220(4): 2087-101, 2015 Jul.
Article in English | MEDLINE | ID: mdl-24777283

ABSTRACT

Astrocytes are highly polarised cells with processes that ensheath microvessels, cover the brain surface, and abut synapses. The endfoot membrane domains facing microvessels and pia are enriched with aquaporin-4 water channels (AQP4) and other members of the dystrophin associated protein complex (DAPC). Several lines of evidence show that loss of astrocyte polarization, defined by the loss of proteins that are normally enriched in astrocyte endfeet, is a common denominator of several neurological diseases such as mesial temporal lobe epilepsy, Alzheimer's disease, and stroke. Little is known about the mechanisms responsible for inducing astrocyte polarization in vivo. Here we introduce the term endfoot-basal lamina junctional complex (EBJC) to denote the proteins that consolidate and characterize the gliovascular interface. The present study was initiated in order to resolve the developmental profile of the EBJC in mouse brain. We show that the EBJC is established after the first week postnatally. Through a combination of methodological approaches, including light microscopic and high resolution immunogold cytochemistry, quantitative RT-PCR, and Western blotting, we demonstrate that the different members of this complex exhibit distinct ontogenic profiles­with the extracellular matrix (ECM) proteins laminin and agrin appearing earlier than the other members of the complex. Specifically, while laminin and agrin expression peak at P7, quantitative immunoblot analyses indicate that AQP4, α-syntrophin, and the inwardly rectifying K(+) channel Kir4.1 expression increases towards adulthood. Our findings are consistent with ECM having an instructive role in establishing astrocyte polarization in postnatal development and emphasize the need to explore the involvement of ECM in neurological disease.


Subject(s)
Astrocytes/physiology , Brain/cytology , Brain/growth & development , Cell Polarity/physiology , Dystrophin-Associated Protein Complex/metabolism , Gene Expression Regulation, Developmental/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Aquaporin 4/genetics , Aquaporin 4/metabolism , Astrocytes/metabolism , Astrocytes/ultrastructure , Dystroglycans/genetics , Dystroglycans/metabolism , Female , Male , Mice, Inbred C57BL , Microscopy, Immunoelectron , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy
19.
Anat Rec (Hoboken) ; 297(9): 1694-705, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25125182

ABSTRACT

The dystrophin-associated glycoprotein complex (DGC) is a collection of glycoproteins that are essential for the normal function of striated muscle and many other tissues. Recent genetic studies have implicated the components of this complex in over a dozen forms of muscular dystrophy. Furthermore, disruption of the DGC has been implicated in many forms of acquired disease. This review aims to summarize the current state of knowledge regarding the processing and assembly of dystrophin-associated proteins with a focus primarily on the dystroglycan heterodimer and the sarcoglycan complex. These proteins form the transmembrane portion of the DGC and undergo a complex multi-step processing with proteolytic cleavage, differential assembly, and both N- and O-glycosylation. The enzymes responsible for this processing and a model describing the sequence and subcellular localization of these events are discussed.


Subject(s)
Dystrophin-Associated Protein Complex/metabolism , Muscle, Striated/metabolism , Protein Processing, Post-Translational , Animals , Dystroglycans/metabolism , Dystrophin-Associated Protein Complex/chemistry , Dystrophin-Associated Protein Complex/genetics , Glycosylation , Humans , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Protein Multimerization , Sarcoglycans/metabolism
20.
J Genet Genomics ; 41(8): 417-28, 2014 Aug 20.
Article in English | MEDLINE | ID: mdl-25160974

ABSTRACT

Myosin XVIIIA, or MYO18A, is a unique PDZ domain-containing unconventional myosin and is evolutionarily conserved from Drosophila to vertebrates. Although there is evidence indicating its expression in the somites, whether it regulates muscle function remains unclear. We show that the two zebrafish myo18a genes (myo18aa and myo18ab) are predominantly expressed at somite borders during early developmental stages. Knockdown of these genes or overexpression of the MYO18A PDZ domain disrupts myofiber integrity, induces myofiber lesions, and compromises the localization of dystrophin, α-dystroglycan (α-DG) and laminin at the myotome boundaries. Cell transplantation experiments indicate that myo18a morphant myoblasts fail to form elongated myofibers in the myotomes of wild-type embryos, which can be rescued by the full-length MYO18A protein. These results suggest that MYO18A likely functions in the adhesion process that maintains the stable attachment of myofibers to ECM (extracellular matrix) and muscle integrity during early development.


Subject(s)
Embryo, Nonmammalian/metabolism , Muscles/embryology , Muscles/metabolism , Myosins/chemistry , Myosins/metabolism , PDZ Domains , Zebrafish/embryology , Animals , Cell Adhesion , Dystroglycans/metabolism , Dystrophin/metabolism , Dystrophin-Associated Protein Complex/metabolism , Embryo, Nonmammalian/cytology , Gene Knockdown Techniques , Lamins/metabolism , Muscles/cytology , Myoblasts/cytology , Myoblasts/metabolism , Myosins/deficiency , Myosins/genetics , Protein Transport , Somites/cytology , Somites/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...