Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 111(49): 17570-5, 2014 Dec 09.
Article in English | MEDLINE | ID: mdl-25422440

ABSTRACT

Misfolded proteins are an emerging hallmark of cardiac diseases. Although some misfolded proteins, such as desmin, are associated with mutations in the genes encoding these disease-associated proteins, little is known regarding more general mechanisms that contribute to the generation of misfolded proteins in the heart. Reduced translational fidelity, caused by a hypomorphic mutation in the editing domain of alanyl-tRNA synthetase (AlaRS), resulted in accumulation of misfolded proteins in specific mouse neurons. By further genetic modulation of the editing activity of AlaRS, we generated mouse models with broader phenotypes, the severity of which was directly related to the degree of compromised editing. Severe disruption of the editing activity of AlaRS caused embryonic lethality, whereas an intermediate reduction in AlaRS editing efficacy resulted in ubiquitinated protein aggregates and mitochondrial defects in cardiomyocytes that were accompanied by progressive cardiac fibrosis and dysfunction. In addition, autophagic vacuoles accumulated in mutant cardiomyocytes, suggesting that autophagy is insufficient to eliminate misfolded proteins. These findings demonstrate that the pathological consequences of diminished tRNA synthetase editing activity, and thus translational infidelity, are dependent on the cell type and the extent of editing disruption, and provide a previously unidentified mechanism underlying cardiac proteinopathy.


Subject(s)
Alanine-tRNA Ligase/deficiency , Alanine-tRNA Ligase/genetics , Heart Diseases/genetics , Proteostasis Deficiencies/genetics , RNA Editing , Alleles , Animals , Bacterial Proteins/genetics , Echocardiography , Homeostasis , Humans , Hydrolysis , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Models, Molecular , Mutation , Myocytes, Cardiac/ultrastructure , Paraffin/chemistry , Protein Binding , Protein Folding , Protein Structure, Tertiary
2.
Acta Neuropathol Commun ; 1: 58, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24252195

ABSTRACT

BACKGROUND: Cobblestone lissencephaly is a severe neuronal migration disorder associated with congenital muscular dystrophies (CMD) such as Walker-Warburg syndrome, muscle-eye-brain disease, and Fukuyama-type CMD. In these severe forms of dystroglycanopathy, the muscular dystrophy and other tissue pathology is caused by mutations in genes involved in O-linked glycosylation of alpha-dystroglycan. While cerebellar dysplasia is a common feature of dystroglycanopathy, its pathogenesis has not been thoroughly investigated. RESULTS: Here we evaluate the role of dystroglycan during cerebellar development. Brain-selective deletion of dystroglycan does not affect overall cerebellar growth, yet causes malformations associated with glia limitans disruptions and granule cell heterotopia that recapitulate phenotypes found in dystroglycanopathy patients. Cerebellar pathology in these mice is not evident until birth even though dystroglycan is lost during the second week of embryogenesis. The severity and spatial distribution of glia limitans disruption, Bergmann glia disorganization, and heterotopia exacerbate during postnatal development. Astrogliosis becomes prominent at these same sites by the time cerebellar development is complete. Interestingly, there is spatial heterogeneity in the glia limitans and granule neuron migration defects that spares the tips of lobules IV-V and VI. CONCLUSIONS: The full spectrum of developmental pathology is caused by loss of dystroglycan from Bergmann glia, as neither granule cell- nor Purkinje cell-specific deletion of dystroglycan results in similar pathology. These data illustrate the importance of dystroglycan function in radial/Bergmann glia, not neurons, for normal cerebellar histogenesis. The spatial heterogeneity of pathology suggests that the dependence on dystroglycan is not uniform.


Subject(s)
Cell Movement/physiology , Cerebellum/growth & development , Cerebellum/physiology , Neuroglia/physiology , Neurons/physiology , Animals , Basement Membrane/metabolism , Bromodeoxyuridine , Cerebellum/ultrastructure , Dystroglycans/genetics , Dystroglycans/metabolism , Fluorescent Antibody Technique , Gliosis/physiopathology , Mice, Knockout , Microscopy, Electron , Neuroglia/ultrastructure
3.
J Neuropathol Exp Neurol ; 71(12): 1047-63, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23147502

ABSTRACT

Interactions between the embryonic pial basement membrane (PBM) and radial glia (RG) are essential for morphogenesis of the cerebral cortex because disrupted interactions cause cobblestone malformations. To elucidate the role of dystroglycan (DG) in PBM-RG interactions, we studied the expression of DG protein and Dag1 mRNA (which encodes DG protein) in developing cerebral cortex and analyzed cortical phenotypes in Dag1 CNS conditional mutant mice. In normal embryonic cortex, Dag1 mRNA was expressed in the ventricular zone, which contains RG nuclei, whereas DG protein was expressed at the cortical surface on RG end feet. Breaches of PBM continuity appeared during early neurogenesis in Dag1 mutants. Diverse cellular elements streamed through the breaches to form leptomeningeal heterotopia that were confluent with the underlying residual cortical plate and contained variably truncated RG fibers, many types of cortical neurons, and radial and intermediate progenitor cells. Nevertheless, layer-specific molecular expression seemed normal in heterotopic neurons, and axons projected to appropriate targets. Dendrites, however, were excessively tortuous and lacked radial orientation. These findings indicate that DG is required on RG end feet to maintain PBM integrity and suggest that cobblestone malformations involve disturbances of RG structure, progenitor distribution, and dendrite orientation, in addition to neuronal "overmigration."


Subject(s)
Basement Membrane , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Dystroglycans/metabolism , Gene Expression Regulation, Developmental/genetics , Neuroglia/cytology , Age Factors , Animals , Basement Membrane/cytology , Basement Membrane/embryology , Basement Membrane/metabolism , Bromodeoxyuridine/metabolism , Cell Movement/genetics , Cell Proliferation , Dystroglycans/genetics , Embryo, Mammalian , Female , In Situ Nick-End Labeling , Intermediate Filament Proteins/deficiency , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Nestin , Neurons/physiology , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Stem Cells/physiology , Tumor Suppressor Proteins/metabolism
4.
Proc Natl Acad Sci U S A ; 108(42): 17426-31, 2011 Oct 18.
Article in English | MEDLINE | ID: mdl-21987822

ABSTRACT

α-dystroglycan is a highly O-glycosylated extracellular matrix receptor that is required for anchoring of the basement membrane to the cell surface and for the entry of Old World arenaviruses into cells. Like-acetylglucosaminyltransferase (LARGE) is a key molecule that binds to the N-terminal domain of α-dystroglycan and attaches ligand-binding moieties to phosphorylated O-mannose on α-dystroglycan. Here we show that the LARGE modification required for laminin- and virus-binding occurs on specific Thr residues located at the extreme N terminus of the mucin-like domain of α-dystroglycan. Deletion and mutation analyses demonstrate that the ligand-binding activity of α-dystroglycan is conferred primarily by LARGE modification at Thr-317 and -319, within the highly conserved first 18 amino acids of the mucin-like domain. The importance of these paired residues in laminin-binding and clustering activity on myoblasts and in arenavirus cell entry is confirmed by mutational analysis with full-length dystroglycan. We further demonstrate that a sequence of five amino acids, Thr(317)ProThr(319)ProVal, contains phosphorylated O-glycosylation and, when modified by LARGE is sufficient for laminin-binding. Because the N-terminal region adjacent to the paired Thr residues is removed during posttranslational maturation of dystroglycan, our results demonstrate that the ligand-binding activity resides at the extreme N terminus of mature α-dystroglycan and is crucial for α-dystroglycan to coordinate the assembly of extracellular matrix proteins and to bind arenaviruses on the cell surface.


Subject(s)
Arenaviridae Infections/etiology , Arenaviridae Infections/metabolism , Dystroglycans/metabolism , Laminin/metabolism , Lymphocytic choriomeningitis virus , N-Acetylglucosaminyltransferases/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Dystroglycans/chemistry , Dystroglycans/genetics , Glycosylation , HEK293 Cells , Humans , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Models, Biological , Molecular Sequence Data , Mutagenesis , Myoblasts/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Threonine/chemistry , Virus Internalization
5.
Eur J Neurosci ; 33(12): 2179-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21501259

ABSTRACT

The dystrophin-dystroglycan complex (DDC) is a molecular array of proteins in muscle and brain cells. The central component of the DDC is dystroglycan, which comprises α- and ß-subunits. α-Dystroglycan (α-DG) binds to extracellular matrix components such as agrin, whereas ß-dystroglycan (ß-DG) is a membrane-spanning protein linking α-DG to the cytoskeleton and other intracellular components such as α-syntrophin. In astrocytes, α-syntrophin binds to the water channel protein aquaporin-4 (AQP4). Recently, it has been shown that AQP4 expression is unaltered in agrin-knockout mice, but that formation of orthogonal arrays of particles (OAPs), consisting of AQP4, is abnormal. As the brain-selective deletion of the DG gene causes a disorganization of the astroglial endfeet, we investigated whether DG deletion has an impact on AQP4. Western blotting revealed reduced AQP4 in the parenchymal but not in the superficial compartment of the astrocyte-conditioned DG-knockout mouse brain. Accordingly, immunohistochemical stainings of AQP4 revealed a selective loss of AQP4 in perivascular but not in superficial astroglial endfeet. In both superficial and perivascular endfeet of the DG-knockout brain, we observed a loss of OAPs. We conclude that in the absence of DG the majority of superficial AQP4 molecules did not form OAPs, and that expression of AQP4 in perivascular endfeet is compromised. However, the decreased number of perivascular AQP4 molecules obviously did form a few OAPs, even in the absence of DG.


Subject(s)
Astrocytes/physiology , Cell Membrane/physiology , Dystroglycans/physiology , Animals , Aquaporin 4/metabolism , Aquaporin 4/physiology , Astrocytes/metabolism , Astrocytes/ultrastructure , Blood-Brain Barrier/physiology , Blood-Brain Barrier/ultrastructure , Brain/metabolism , Brain/physiology , Brain/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Dystroglycans/genetics , Freeze Fracturing/methods , Mice , Mice, Knockout
6.
J Biol Chem ; 286(24): 21180-90, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21460210

ABSTRACT

Dystroglycanopathies are a subset of congenital muscular dystrophies wherein α-dystroglycan (α-DG) is hypoglycosylated. α-DG is an extensively O-glycosylated extracellular matrix-binding protein and a key component of the dystrophin-glycoprotein complex. Previous studies have shown α-DG to be post-translationally modified by both O-GalNAc- and O-mannose-initiated glycan structures. Mutations in defined or putative glycosyltransferase genes involved in O-mannosylation are associated with a loss of ligand-binding activity of α-DG and are causal for various forms of congenital muscular dystrophy. In this study, we sought to perform glycomic analysis on brain O-linked glycan structures released from proteins of three different knock-out mouse models associated with O-mannosylation (POMGnT1, LARGE (Myd), and DAG1(-/-)). Using mass spectrometry approaches, we were able to identify nine O-mannose-initiated and 25 O-GalNAc-initiated glycan structures in wild-type littermate control mouse brains. Through our analysis, we were able to confirm that POMGnT1 is essential for the extension of all observed O-mannose glycan structures with ß1,2-linked GlcNAc. Loss of LARGE expression in the Myd mouse had no observable effect on the O-mannose-initiated glycan structures characterized here. Interestingly, we also determined that similar amounts of O-mannose-initiated glycan structures are present on brain proteins from α-DG-lacking mice (DAG1) compared with wild-type mice, indicating that there must be additional proteins that are O-mannosylated in the mammalian brain. Our findings illustrate that classical ß1,2-elongation and ß1,6-GlcNAc branching of O-mannose glycan structures are dependent upon the POMGnT1 enzyme and that O-mannosylation is not limited solely to α-DG in the brain.


Subject(s)
Glycomics/methods , Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/genetics , Animals , Brain/metabolism , Carbohydrates/chemistry , Disease Models, Animal , Dystroglycans/chemistry , Galactosyltransferases/chemistry , Glycosylation , Mannose/chemistry , Mice , Mice, Knockout , Muscular Dystrophies/congenital , Mutation , Polysaccharides/chemistry
7.
N Engl J Med ; 364(10): 939-46, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21388311

ABSTRACT

Dystroglycan, which serves as a major extracellular matrix receptor in muscle and the central nervous system, requires extensive O-glycosylation to function. We identified a dystroglycan missense mutation (Thr192→Met) in a woman with limb-girdle muscular dystrophy and cognitive impairment. A mouse model harboring this mutation recapitulates the immunohistochemical and neuromuscular abnormalities observed in the patient. In vitro and in vivo studies showed that the mutation impairs the receptor function of dystroglycan in skeletal muscle and brain by inhibiting the post-translational modification, mediated by the glycosyltransferase LARGE, of the phosphorylated O-mannosyl glycans on α-dystroglycan that is required for high-affinity binding to laminin.


Subject(s)
Dystroglycans/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Mutation, Missense , Animals , Disease Models, Animal , Female , Humans , Mice , Pedigree , Phenotype , Sequence Analysis, DNA
8.
J Neurosci ; 30(43): 14560-72, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20980614

ABSTRACT

Cobblestone (type II) lissencephaly and mental retardation are characteristic features of a subset of congenital muscular dystrophies that include Walker-Warburg syndrome, muscle-eye-brain disease, and Fukuyama-type congenital muscular dystrophy. Although the majority of clinical cases are genetically undefined, several causative genes have been identified that encode known or putative glycosyltransferases in the biosynthetic pathway of dystroglycan. Here we test the effects of brain-specific deletion of dystroglycan, and show distinct functions for neuronal and glial dystroglycan. Deletion of dystroglycan in the whole brain produced glial/neuronal heterotopia resembling the cerebral cortex malformation in cobblestone lissencephaly. In wild-type mice, dystroglycan stabilizes the basement membrane of the glia limitans, thereby supporting the cortical infrastructure necessary for neuronal migration. This function depends on extracellular dystroglycan interactions, since the cerebral cortex developed normally in transgenic mice that lack the dystroglycan intracellular domain. Also, forebrain histogenesis was preserved in mice with neuron-specific deletion of dystroglycan, but hippocampal long-term potentiation was blunted, as is also the case in the Largemyd mouse, in which dystroglycan glycosylation is disrupted. Our findings provide genetic evidence that neuronal dystroglycan plays a role in synaptic plasticity and that glial dystroglycan is involved in forebrain development. Differences in dystroglycan glycosylation in distinct cell types of the CNS may contribute to the diversity of dystroglycan function in the CNS, as well as to the broad clinical spectrum of type II lissencephalies.


Subject(s)
Brain/growth & development , Brain/physiology , Dystroglycans/physiology , Neuroglia/physiology , Neurons/physiology , Animals , Brain/abnormalities , Brain Chemistry/genetics , Brain Chemistry/physiology , Dystroglycans/genetics , Dystroglycans/metabolism , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Fluorescent Antibody Technique , Genes, myc/genetics , Glial Fibrillary Acidic Protein/genetics , Hippocampus/physiology , Hydrocephalus/genetics , Hydrocephalus/pathology , Intermediate Filament Proteins/genetics , Long-Term Potentiation/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nestin , Neuroglia/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Reverse Transcriptase Polymerase Chain Reaction
9.
J Neurosci ; 29(42): 13136-46, 2009 Oct 21.
Article in English | MEDLINE | ID: mdl-19846701

ABSTRACT

Ocular involvement in muscular dystrophy ranges from structural defects to abnormal electroretinograms. While the mechanisms underlying the abnormal retinal physiology in patients are not understood, it is thought that alpha-dystroglycan extracellular interactions are critical for normal visual function. Here we show that beta-dystroglycan anchors dystrophin and the inward rectifying K(+) channel Kir4.1 at glial endfeet and that disruption of dystrophin and potassium channel clustering in dystroglycan mutant mice is associated with an attenuation of the electroretinogram b-wave. Glial-specific inactivation of dystroglycan or deletion of the cytoplasmic domain of beta-dystroglycan was sufficient to attenuate the electroretinogram b-wave. Unexpectedly, deletion of the beta-dystroglycan cytoplasmic domain did not disrupt the laminar structure of the retina. In contrast to the role of alpha-dystroglycan extracellular interactions during early development of the CNS, beta-dystroglycan intracellular interactions are important for visual function but not the laminar development of the retina.


Subject(s)
Dystroglycans/deficiency , Vision Disorders/genetics , Vision Disorders/physiopathology , Animals , Dystrophin/metabolism , Electroretinography/methods , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/metabolism , Laminin/genetics , Laminin/metabolism , Maze Learning/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nestin , Photic Stimulation/methods , Potassium Channels, Inwardly Rectifying/metabolism , Retina/metabolism , Retina/pathology , Visual Fields/genetics
10.
J Neurosci ; 28(42): 10567-75, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18923033

ABSTRACT

Walker-Warburg syndrome (WWS) is a severe congenital disease that is characterized by brain and eye malformations and lethality during the first year of life. Genetic mutations have been identified in a subset of WWS patients, but a majority of clinical cases have unknown etiologies. POMT1 and POMT2, two of the causative genes, form an active enzyme complex in the posttranslational biosynthetic pathway of dystroglycan. Deletion of either Pomt1 or the dystroglycan gene causes early embryonic lethality in mice. Here we report that mice with epiblast-specific loss of dystroglycan develop brain and eye defects that broadly resemble the clinical spectrum of the human disease, including aberrant neuron migration, hydrocephalus, and malformations of the anterior and posterior chambers of the eye. Breaches of basement membranes coincide with the pathology, revealing an important function for dystroglycan in the morphogenesis of the brain and eye. These findings demonstrate the central role of dystroglycan in WWS and suggest that novel defects in posttranslational processing or mutations of the dystroglycan gene itself may underlie cases in which no causative mutation has been found.


Subject(s)
Brain/abnormalities , Brain/metabolism , Dystroglycans/deficiency , Eye Abnormalities/metabolism , Germ Layers/abnormalities , Germ Layers/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Animals , Brain/pathology , Dystroglycans/genetics , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Germ Layers/pathology , Humans , Hydrocephalus/genetics , Hydrocephalus/metabolism , Hydrocephalus/pathology , Infant , Male , Mice , Mice, Knockout , Mice, Transgenic , Syndrome
12.
FEBS Lett ; 579(21): 4792-6, 2005 Aug 29.
Article in English | MEDLINE | ID: mdl-16098969

ABSTRACT

Dystroglycan is a cell-surface matrix receptor that requires LARGE-dependent glycosylation for laminin binding. Although the interaction of dystroglycan with laminin has been well characterized, less is known about the role of dystroglycan glycosylation in the binding and assembly of perlecan. We report reduced perlecan-binding activity and mislocalization of perlecan in the LARGE-deficient Large(myd) mouse. Cell-surface ligand clustering assays show that laminin polymerization promotes perlecan assembly. Solid-phase binding assays provide evidence for the first time of a trimolecular complex formation of dystroglycan, laminin and perlecan. These data suggest functional disruption of the trimolecular complex in glycosylation-deficient muscular dystrophy.


Subject(s)
Dystroglycans , Extracellular Matrix/metabolism , Heparan Sulfate Proteoglycans/metabolism , Animals , Antibodies, Monoclonal/metabolism , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Humans , Laminin/metabolism , Mice , Mice, Inbred C57BL , Multiprotein Complexes , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational
13.
Glia ; 49(4): 492-500, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15578661

ABSTRACT

We investigate the role of dystroglycan, a major laminin-1 receptor and central member of the dystrophin-glycoprotein complex, in the laminin-1 induced motility of cultured Muller glial cells. Binding of laminin-1 to dystroglycan was prevented by IIH6, a function-blocking monoclonal antibody against alpha-dystroglycan. As an alternative means of inhibition, we used heparin to mask the dystroglycan binding site of the laminin-1, known to overlap with heparin binding sites. Cell motility was characterized in a two-dimensional motility assay based on computer-controlled videomicroscopy and statistical analysis of cellular trajectories. We obtained data on both the cell velocity and the diffusion index, a measure of direction-changing frequency. Both means of inhibition of dystroglycan function led to a significant decrease in the ability of laminin-1 to stimulate cell migration. At the same time, dystroglycan function does not appear to be involved in laminin-1-dependent increase in process dynamism and direction-changing activity.


Subject(s)
Cell Movement/physiology , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Laminin/metabolism , Neuroglia/metabolism , Retina/metabolism , Animals , Animals, Newborn , Antibodies/pharmacology , Binding Sites/drug effects , Binding Sites/physiology , Cell Movement/drug effects , Cells, Cultured , Heparin/pharmacology , Laminin/pharmacology , Microscopy, Video , Neuroglia/cytology , Neuroglia/drug effects , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Wistar , Retina/cytology , Retina/growth & development , Up-Regulation/drug effects , Up-Regulation/physiology
14.
Nat Med ; 10(7): 696-703, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15184894

ABSTRACT

Several congenital muscular dystrophies caused by defects in known or putative glycosyltransferases are commonly associated with hypoglycosylation of alpha-dystroglycan (alpha-DG) and a marked reduction of its receptor function. We have investigated changes in the processing and function of alpha-DG resulting from genetic manipulation of LARGE, the putative glycosyltransferase mutated both in Large(myd) mice and in humans with congenital muscular dystrophy 1D (MDC1D). Here we show that overexpression of LARGE ameliorates the dystrophic phenotype of Large(myd) mice and induces the synthesis of glycan-enriched alpha-DG with high affinity for extracellular ligands. Notably, LARGE circumvents the alpha-DG glycosylation defect in cells from individuals with genetically distinct types of congenital muscular dystrophy. Gene transfer of LARGE into the cells of individuals with congenital muscular dystrophies restores alpha-DG receptor function, whereby glycan-enriched alpha-DG coordinates the organization of laminin on the cell surface. Our findings indicate that modulation of LARGE expression or activity is a viable therapeutic strategy for glycosyltransferase-deficient congenital muscular dystrophies.


Subject(s)
Cytoskeletal Proteins/metabolism , Glycosyltransferases/deficiency , Membrane Glycoproteins/metabolism , Muscular Dystrophies/congenital , N-Acetylglucosaminyltransferases/physiology , Neoplasm Proteins/physiology , Animals , Dystroglycans , Genetic Therapy , Glycosylation , Humans , Laminin/metabolism , Mice , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , N-Acetylglucosaminyltransferases/genetics , Neoplasm Proteins/genetics
15.
Nature ; 418(6896): 417-22, 2002 Jul 25.
Article in English | MEDLINE | ID: mdl-12140558

ABSTRACT

Muscle eye brain disease (MEB) and Fukuyama congenital muscular dystrophy (FCMD) are congenital muscular dystrophies with associated, similar brain malformations. The FCMD gene, fukutin, shares some homology with fringe-like glycosyltransferases, and the MEB gene, POMGnT1, seems to be a new glycosyltransferase. Here we show, in both MEB and FCMD patients, that alpha-dystroglycan is expressed at the muscle membrane, but similar hypoglycosylation in the diseases directly abolishes binding activity of dystroglycan for the ligands laminin, neurexin and agrin. We show that this post-translational biochemical and functional disruption of alpha-dystroglycan is recapitulated in the muscle and central nervous system of mutant myodystrophy (myd) mice. We demonstrate that myd mice have abnormal neuronal migration in cerebral cortex, cerebellum and hippocampus, and show disruption of the basal lamina. In addition, myd mice reveal that dystroglycan targets proteins to functional sites in brain through its interactions with extracellular matrix proteins. These results suggest that at least three distinct mammalian genes function within a convergent post-translational processing pathway during the biosynthesis of dystroglycan, and that abnormal dystroglycan-ligand interactions underlie the pathogenic mechanism of muscular dystrophy with brain abnormalities.


Subject(s)
Cytoskeletal Proteins/metabolism , Membrane Glycoproteins/metabolism , Muscular Dystrophies/congenital , Muscular Dystrophies/metabolism , Protein Processing, Post-Translational , Animals , Brain/abnormalities , Brain/metabolism , Brain/pathology , Cell Movement , Cytoskeletal Proteins/genetics , Dystroglycans , Extracellular Matrix Proteins/metabolism , Eye/metabolism , Female , Gene Deletion , Glycosylation , Humans , Ligands , Male , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Muscles/metabolism , Neurons/metabolism , Neurons/pathology , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...