Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
FASEB Bioadv ; 5(11): 453-469, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37936920

ABSTRACT

Store-operated Ca2+ entry (SOCE) is indispensable for intracellular Ca2+ homeostasis in skeletal muscle, and constitutive activation of SOCE causes tubular aggregate myopathy (TAM). To understand the pathogenesis of TAM, we induced pluripotent stem cells (iPSCs) from a TAM patient with a rare mutation (c.1450_1451insGA; p. Ile484ArgfsX21) in the STIM1 gene. This frameshift mutation produces a truncated STIM1 with a disrupted C-terminal inhibitory domain (CTID) and was reported to diminish SOCE. Myotubes induced from the patient's-iPSCs (TAM myotubes) showed severely impaired SOCE, but antioxidants greatly restored SOCE partly via upregulation of an endoplasmic reticulum (ER) chaperone, BiP (GRP78), in the TAM myotubes. Our observation suggests that antioxidants are promising tools for treatment of TAM caused by reduced SOCE.

2.
Elife ; 122023 02 01.
Article in English | MEDLINE | ID: mdl-36723429

ABSTRACT

Dystroglycan (DG) requires extensive post-translational processing and O-glycosylation to function as a receptor for extracellular matrix (ECM) proteins containing laminin-G (LG) domains. Matriglycan is an elongated polysaccharide of alternating xylose (Xyl) and glucuronic acid (GlcA) that binds with high affinity to ECM proteins with LG domains and is uniquely synthesized on α-dystroglycan (α-DG) by like-acetylglucosaminyltransferase-1 (LARGE1). Defects in the post-translational processing or O-glycosylation of α-DG that result in a shorter form of matriglycan reduce the size of α-DG and decrease laminin binding, leading to various forms of muscular dystrophy. Previously, we demonstrated that protein O-mannose kinase (POMK) is required for LARGE1 to generate full-length matriglycan on α-DG (~150-250 kDa) (Walimbe et al., 2020). Here, we show that LARGE1 can only synthesize a short, non-elongated form of matriglycan in mouse skeletal muscle that lacks the DG N-terminus (α-DGN), resulting in an ~100-125 kDa α-DG. This smaller form of α-DG binds laminin and maintains specific force but does not prevent muscle pathophysiology, including reduced force production after eccentric contractions (ECs) or abnormalities in the neuromuscular junctions. Collectively, our study demonstrates that α-DGN, like POMK, is required for LARGE1 to extend matriglycan to its full mature length on α-DG and thus prevent muscle pathophysiology.


Subject(s)
Dystroglycans , Muscular Dystrophies , N-Acetylglucosaminyltransferases , Animals , Mice , Dystroglycans/metabolism , Extracellular Matrix Proteins/metabolism , Glycosylation , Laminin/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Protein Kinases/metabolism , Protein Processing, Post-Translational , N-Acetylglucosaminyltransferases/metabolism
3.
Mol Ther Methods Clin Dev ; 18: 131-144, 2020 Sep 11.
Article in English | MEDLINE | ID: mdl-32637445

ABSTRACT

Myotonic dystrophy type 1 is the most common type of adult-onset muscular dystrophy. This is an autosomal dominant disorder and caused by the expansion of the CTG repeat in the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. Messenger RNAs containing these expanded repeats form aggregates as nuclear RNA foci. Then, RNA binding proteins, including muscleblind-like 1, are sequestered to the RNA foci, leading to systemic abnormal RNA splicing. In this study, we used CRISPR-Cas9 genome editing to excise this CTG repeat. Dual cleavage at the 5' and 3' regions of the repeat using a conventional Cas9 nuclease and a double nicking with Cas9 nickase successfully excised the CTG repeat. Subsequently, the formation of the RNA foci was markedly reduced in patient-derived fibroblasts. However, contrary to expectations, a considerable amount of off-target digestions and on-target genomic rearrangements were observed using high-throughput genome-wide translocation sequencing. Finally, the suppression of DMPK transcripts using CRISPR interference significantly decreased the intensity of RNA foci. Our results indicate that close attention should be paid to the unintended mutations when double-strand breaks are generated by CRISPR-Cas9 for therapeutic purposes. Alternative approaches independent of double-strand breaks, including CRISPR interference, may be considered.

4.
Proc Natl Acad Sci U S A ; 116(23): 11396-11401, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31097590

ABSTRACT

α-Dystroglycan (α-DG) is a highly glycosylated basement membrane receptor that is cleaved by the proprotein convertase furin, which releases its N-terminal domain (α-DGN). Before cleavage, α-DGN interacts with the glycosyltransferase LARGE1 and initiates functional O-glycosylation of the mucin-like domain of α-DG. Notably, α-DGN has been detected in a wide variety of human bodily fluids, but the physiological significance of secreted α-DGN remains unknown. Here, we show that mice lacking α-DGN exhibit significantly higher viral titers in the lungs after Influenza A virus (IAV) infection (strain A/Puerto Rico/8/1934 H1N1), suggesting an inability to control virus load. Consistent with this, overexpression of α-DGN before infection or intranasal treatment with recombinant α-DGN prior and during infection, significantly reduced IAV titers in the lungs of wild-type mice. Hemagglutination inhibition assays using recombinant α-DGN showed in vitro neutralization of IAV. Collectively, our results support a protective role for α-DGN in IAV proliferation.


Subject(s)
Cell Proliferation/drug effects , Dystroglycans/pharmacology , Influenza A Virus, H1N1 Subtype/drug effects , Protective Agents/pharmacology , Animals , Basement Membrane/drug effects , Basement Membrane/virology , Body Fluids/drug effects , Body Fluids/virology , Cell Line , Glycosylation/drug effects , HEK293 Cells , Humans , Inflammation/drug therapy , Inflammation/virology , Influenza, Human/drug therapy , Influenza, Human/virology , Lung/drug effects , Lung/virology , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Viral Load/methods
5.
Proc Natl Acad Sci U S A ; 113(39): 10992-7, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27625424

ABSTRACT

Dystroglycan (DG) is a highly expressed extracellular matrix receptor that is linked to the cytoskeleton in skeletal muscle. DG is critical for the function of skeletal muscle, and muscle with primary defects in the expression and/or function of DG throughout development has many pathological features and a severe muscular dystrophy phenotype. In addition, reduction in DG at the sarcolemma is a common feature in muscle biopsies from patients with various types of muscular dystrophy. However, the consequence of disrupting DG in mature muscle is not known. Here, we investigated muscles of transgenic mice several months after genetic knockdown of DG at maturity. In our study, an increase in susceptibility to contraction-induced injury was the first pathological feature observed after the levels of DG at the sarcolemma were reduced. The contraction-induced injury was not accompanied by increased necrosis, excitation-contraction uncoupling, or fragility of the sarcolemma. Rather, disruption of the sarcomeric cytoskeleton was evident as reduced passive tension and decreased titin immunostaining. These results reveal a role for DG in maintaining the stability of the sarcomeric cytoskeleton during contraction and provide mechanistic insight into the cause of the reduction in strength that occurs in muscular dystrophy after lengthening contractions.


Subject(s)
Cytoskeleton/metabolism , Dystroglycans/metabolism , Muscle Contraction , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Sarcomeres/metabolism , Animals , Connectin/metabolism , Cytoskeleton/drug effects , Excitation Contraction Coupling/drug effects , Female , Isometric Contraction/drug effects , Male , Mice, Knockout , Muscle Contraction/drug effects , Muscle, Skeletal/drug effects , Necrosis , Organ Size , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcolemma/metabolism , Sarcomeres/drug effects , Tamoxifen/pharmacology
6.
Neurol Genet ; 2(1): e50, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27066587

ABSTRACT

OBJECTIVE: To identify the gene mutation of tubular aggregate myopathy (TAM) and gain mechanistic insight into the pathogenesis of the disorder. METHODS: We described a family affected by autosomal dominant TAM and performed exome and Sanger sequencing to identify mutations. We further analyzed the functional significance of the identified mutation by expression studies and intracellular Ca(2+) measurements. RESULTS: A 42-year-old man presented with slowly progressive muscle weakness and atrophy in all 4 limbs and the trunk. Muscle biopsy and microscopic examination revealed tubular aggregates in his skeletal muscle. Genetic analysis of this family identified a novel heterozygous mutation, c.1450_1451insGA (p.Ile484ArgfsX21), in stromal interaction molecule 1 (STIM1), a Ca(2+) sensor in sarcoplasmic reticulum. We transfected cultured cells with STIM1 and demonstrated that the mutant STIM1 exhibited aggregation-like appearance in shrunk cytoplasm. Furthermore, we revealed that the intracellular Ca(2+) influx is decreased by the mutant STIM1. CONCLUSIONS: The novel mutation p.Ile484ArgfsX21 is located in the cytoplasmic C-terminal inhibitory domain (CTID) of STIM1. However, all mutations reported so far in TAM reside in the luminal N-terminal EF hand region. The aggregation-like appearance of STIM1 and the decreased intracellular Ca(2+) influx in cells transfected with CTID mutant are in sharp contrast to these previous reports. Taken together, these findings indicate that mutations of STIM1 cause TAM through the dysregulation of Ca(2+) homeostasis.

7.
Hum Mol Genet ; 23(17): 4543-58, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24722207

ABSTRACT

Several types of muscular dystrophy are caused by defective linkage between α-dystroglycan (α-DG) and laminin. Among these, dystroglycanopathy, including Fukuyama-type congenital muscular dystrophy (FCMD), results from abnormal glycosylation of α-DG. Recent studies have shown that like-acetylglucosaminyltransferase (LARGE) strongly enhances the laminin-binding activity of α-DG. Therefore, restoration of the α-DG-laminin linkage by LARGE is considered one of the most promising possible therapies for muscular dystrophy. In this study, we generated transgenic mice that overexpress LARGE (LARGE Tg) and crossed them with dy(2J) mice and fukutin conditional knockout mice, a model for laminin α2-deficient congenital muscular dystrophy (MDC1A) and FCMD, respectively. Remarkably, in both the strains, the transgenic overexpression of LARGE resulted in an aggravation of muscular dystrophy. Using morphometric analyses, we found that the deterioration of muscle pathology was caused by suppression of muscle regeneration. Overexpression of LARGE in C2C12 cells further demonstrated defects in myotube formation. Interestingly, a decreased expression of insulin-like growth factor 1 (IGF-1) was identified in both LARGE Tg mice and LARGE-overexpressing C2C12 myotubes. Supplementing the C2C12 cells with IGF-1 restored the defective myotube formation. Taken together, our findings indicate that the overexpression of LARGE aggravates muscular dystrophy by suppressing the muscle regeneration and this adverse effect is mediated via reduced expression of IGF-1.


Subject(s)
Down-Regulation , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/physiopathology , N-Acetylglucosaminyltransferases/metabolism , Regeneration , Animals , Cell Fusion , Cell Line , Dystroglycans/metabolism , Glycosylation , Humans , Mice , Mice, Transgenic , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Myoblasts/metabolism , Myoblasts/pathology , Phenotype , Proteins/metabolism , Transfection , Transferases
8.
Nature ; 503(7474): 136-40, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24132234

ABSTRACT

The dense glycan coat that surrounds every cell is essential for cellular development and physiological function, and it is becoming appreciated that its composition is highly dynamic. Post-translational addition of the polysaccharide repeating unit [-3-xylose-α1,3-glucuronic acid-ß1-]n by like-acetylglucosaminyltransferase (LARGE) is required for the glycoprotein dystroglycan to function as a receptor for proteins in the extracellular matrix. Reductions in the amount of [-3-xylose-α1,3-glucuronic acid-ß1-]n (hereafter referred to as LARGE-glycan) on dystroglycan result in heterogeneous forms of muscular dystrophy. However, neither patient nor mouse studies has revealed a clear correlation between glycosylation status and phenotype. This disparity can be attributed to our lack of knowledge of the cellular function of the LARGE-glycan repeat. Here we show that coordinated upregulation of Large and dystroglycan in differentiating mouse muscle facilitates rapid extension of LARGE-glycan repeat chains. Using synthesized LARGE-glycan repeats we show a direct correlation between LARGE-glycan extension and its binding capacity for extracellular matrix ligands. Blocking Large upregulation during muscle regeneration results in the synthesis of dystroglycan with minimal LARGE-glycan repeats in association with a less compact basement membrane, immature neuromuscular junctions and dysfunctional muscle predisposed to dystrophy. This was consistent with the finding that patients with increased clinical severity of disease have fewer LARGE-glycan repeats. Our results reveal that the LARGE-glycan of dystroglycan serves as a tunable extracellular matrix protein scaffold, the extension of which is required for normal skeletal muscle function.


Subject(s)
Dystroglycans/chemistry , Dystroglycans/metabolism , Extracellular Matrix/metabolism , Muscular Dystrophies/prevention & control , N-Acetylglucosaminyltransferases/metabolism , Polysaccharides/metabolism , Animals , Basement Membrane/metabolism , Basement Membrane/pathology , Cell Differentiation , Cell Line , Extracellular Matrix/chemistry , Female , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Molecular Weight , Muscle Development , Muscles/metabolism , Muscles/pathology , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Myoblasts , N-Acetylglucosaminyltransferases/deficiency , N-Acetylglucosaminyltransferases/genetics , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Phenotype , Polysaccharides/chemistry
9.
Biochem Biophys Res Commun ; 414(4): 826-31, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22019851

ABSTRACT

Histone deacetylase inhibitors (HDACIs) are known to promote skeletal muscle formation. However, their mechanisms that include effects on the expression of major muscle components such as the dystrophin-associated proteins complex (DAPC) or myogenic regulatory factors (MRFs) remain unknown. In this study, we investigated the effects of HDACIs on skeletal muscle formation using the C2C12 cell culture system. C2C12 myoblasts were exposed to trichostatin A (TSA), one of the most potent HDACIs, and differentiation was subsequently induced. We found that TSA enhances the expression of myosin heavy chain without affecting DAPC expression. In addition, TSA increases the expression of the early MRFs, Myf5 and MEF2, whereas it suppresses the expression of the late MRF, myogenin. Interestingly, TSA also enhances the expression of Id1, Id2, and Id3 (Ids). Ids are myogenic repressors that inhibit myogenic differentiation. These findings suggest that TSA promotes gene expression in proliferation and suppresses it in the differentiation stage of muscle formation. Taken together, our data demonstrate that TSA enhances myogenesis by coordinating the expression of MRFs and myogenic repressors.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Myogenic Regulatory Factors/metabolism , Animals , Cell Line , Dystrophin-Associated Protein Complex/metabolism , Gene Expression/drug effects , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Protein 2/metabolism , Inhibitor of Differentiation Proteins/metabolism , Mice , Muscle Development/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Myogenic Regulatory Factor 5/metabolism , Myosin Heavy Chains/metabolism
10.
Biochem Biophys Res Commun ; 411(2): 365-9, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21741360

ABSTRACT

α-Dystroglycan (α-DG) plays crucial roles in maintaining the stability of cells. We demonstrated previously that the N-terminal domain of α-DG (α-DG-N) is secreted by cultured cells into the culture medium. In the present study, to clarify its function in vivo, we generated a monoclonal antibody against α-DG-N and investigated the secretion of α-DG-N in human cerebrospinal fluid (CSF). Interestingly, we found that a considerable amount of α-DG-N was present in CSF. α-DG-N in CSF was a sialylated glycoprotein with both N- and O-linked glycan. These observations suggest that secreted α-DG-N may be transported via CSF and have yet unidentified effects on the nervous system.


Subject(s)
Dystroglycans/cerebrospinal fluid , Dystroglycans/metabolism , Glycoproteins/cerebrospinal fluid , Glycoproteins/metabolism , Neurons/metabolism , Cells, Cultured , Dystroglycans/chemistry , Glycoproteins/chemistry , Glycosylation , Humans , N-Acetylneuraminic Acid/chemistry , Protein Structure, Tertiary
11.
Kidney Int ; 79(3): 311-6, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20944549

ABSTRACT

In addition to skeletal muscle and the nervous system, α-dystroglycan is found in the podocyte basal membrane, stabilizing these cells on the glomerular basement membrane. Fukutin, named after the gene responsible for Fukuyama-type congenital muscular dystrophy, is a putative glycosyltransferase required for the post-translational modification of α-dystroglycan. Chimeric mice targeted for both alleles of fukutin develop severe muscular dystrophy; however, these mice do not have proteinuria. Despite the lack of a functional renal defect, we evaluated glomerular structure and found minor abnormalities in the chimeric mice by light microscopy. Electron microscopy revealed flattening of podocyte foot processes, the number of which was significantly lower in the chimeric compared to wild-type mice. A monoclonal antibody against the laminin-binding carbohydrate residues of α-dystroglycan did not detect α-dystroglycan glycosylation in the glomeruli by immunoblotting or immunohistochemistry. In contrast, expression of the core α-dystroglycan protein was preserved. There was no statistical difference in dystroglycan mRNA expression or in the amount of nephrin and α3-integrin protein in the chimeric compared to the wild-type mice as judged by immunohistochemistry and real-time RT-PCR. Thus, our results indicate that appropriate glycosylation of α-dystroglycan has an important role in the maintenance of podocyte architecture.


Subject(s)
Cell Shape , Dystroglycans/metabolism , Podocytes/metabolism , Protein Processing, Post-Translational , Walker-Warburg Syndrome/metabolism , Animals , Blotting, Western , Disease Models, Animal , Dystroglycans/genetics , Glycosylation , Immunohistochemistry , Integrin alpha3/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Podocytes/pathology , Polymerase Chain Reaction , Proteins/genetics , Proteins/metabolism , RNA, Messenger/metabolism , Transferases , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/pathology
12.
J Biomed Biotechnol ; 2010: 740403, 2010.
Article in English | MEDLINE | ID: mdl-20625412

ABSTRACT

Dystroglycan is a central component of the dystrophin-glycoprotein complex (DGC) that links extracellular matrix with cytoskeleton, expressed in a variety of fetal and adult tissues. Dystroglycan plays diverse roles in development and homeostasis including basement membrane formation, epithelial morphogenesis, membrane stability, cell polarization, and cell migration. In this paper, we will focus on biological role of dystroglycan in Schwann cell function, especially myelination. First, we review the molecular architecture of DGC in Schwann cell abaxonal membrane. Then, we will review the loss-of-function studies using targeted mutagenesis, which have revealed biological functions of each component of DGC in Schwann cells. Based on these findings, roles of dystroglycan in Schwann cell function, in myelination in particular, and its implications in diseases will be discussed in detail. Finally, in view of the fact that understanding the role of dystroglycan in Schwann cells is just beginning, future perspectives will be discussed.


Subject(s)
Dystroglycans/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Schwann Cells/metabolism , Humans , Myelin Sheath/metabolism , Schwann Cells/pathology
13.
Mech Dev ; 126(3-4): 107-16, 2009.
Article in English | MEDLINE | ID: mdl-19114101

ABSTRACT

Protein O-linked mannose beta1,2-N-acetylglucosaminyltransferase 1 (POMGnT1) is an enzyme that transfers N-acetylglucosamine to O-mannose of glycoproteins. Mutations of the POMGnT1 gene cause muscle-eye-brain (MEB) disease. To obtain a better understanding of the pathogenesis of MEB disease, we mutated the POMGnT1 gene in mice using a targeting technique. The mutant muscle showed aberrant glycosylation of alpha-DG, and alpha-DG from mutant muscle failed to bind laminin in a binding assay. POMGnT1(-/-) muscle showed minimal pathological changes with very low-serum creatine kinase levels, and had normally formed muscle basal lamina, but showed reduced muscle mass, reduced numbers of muscle fibers, and impaired muscle regeneration. Importantly, POMGnT1(-/-) satellite cells proliferated slowly, but efficiently differentiated into multinuclear myotubes in vitro. Transfer of a retrovirus vector-mediated POMGnT1 gene into POMGnT1(-/-) myoblasts completely restored the glycosylation of alpha-DG, but proliferation of the cells was not improved. Our results suggest that proper glycosylation of alpha-DG is important for maintenance of the proliferative activity of satellite cells in vivo.


Subject(s)
Myoblasts/cytology , Myoblasts/enzymology , N-Acetylglucosaminyltransferases/deficiency , Animals , Cell Proliferation , Cells, Cultured , Creatine Kinase/blood , Embryonic Stem Cells/metabolism , Fibrosis/complications , Fibrosis/enzymology , Fibrosis/pathology , Gene Deletion , Gene Targeting , Immunohistochemistry , Mice , Muscle, Skeletal/enzymology , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Muscular Dystrophy, Animal/blood , Muscular Dystrophy, Animal/complications , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Animal/pathology , Myoblasts/ultrastructure , N-Acetylglucosaminyltransferases/metabolism , Phenotype , Regeneration , Satellite Cells, Skeletal Muscle/enzymology , Satellite Cells, Skeletal Muscle/pathology , Satellite Cells, Skeletal Muscle/ultrastructure , Signal Transduction
14.
J Neuroimaging ; 19(3): 295-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-18482367

ABSTRACT

Primary non-Hodgkin lymphoma of the skull base is a rare disorder. We report a case of primary non-Hodgkin lymphoma of the skull base presenting with Garcin syndrome. MRI revealed peculiar lesions in the cavernous sinus, clivus, and occipital bone. Diagnosis was made by biopsy of the tumor in the cavernous sinus.


Subject(s)
Cranial Nerve Diseases/pathology , Lymphoma, Non-Hodgkin/pathology , Skull Neoplasms/pathology , Aged , Cranial Nerve Diseases/diagnosis , Diagnosis, Differential , Humans , Lymphoma, Non-Hodgkin/diagnosis , Magnetic Resonance Imaging , Male , Skull/pathology , Skull Neoplasms/diagnosis , Syndrome
15.
Rinsho Shinkeigaku ; 48(8): 543-9, 2008 Aug.
Article in Japanese | MEDLINE | ID: mdl-18939472

ABSTRACT

Congenital muscular dystrophy (CMD) refers to a heterogeneous group of muscular dystrophies with onset during the neonatal period. Among them, some types of CMD are characterized by the association of brain malformations and ocular abnormalities. Biochemical analyses revealed altered glycosylation and decreased laminin-binding activity of alpha-dystroglycan in these disorders, therefore they are correctively called alpha-dystroglycanopathy. Recently, mutations in the genes encoding demonstrated or putative glycosyltransferases have been identified in alpha-dystroglycanopathy. Fukuyama-type CMD and MDC1C are caused by mutations in the fukutin and fukutin-related protein (FKRP) genes, respectively. Mutations in the protein O-mannose beta-1, 2-N-acetylglucosaminyltransferase (POMGnT-1) and protein O-mannosyltransferase 1 and 2 (POMT1 and POMT2) genes cause muscle-eye-brain disease and Walker-Warburg syndrome, respectively. In addition, mutations in Large gene results in MDC1D. Furthermore, recent genotype-phenotype correlation analyses have revealed that the spectrum of phenotypes caused by mutations in these genes is much wider than originally assumed. In this review, we focus on the molecular pathomechanism and diverging clinical phenotypes of alpha-dystroglycanopathy.


Subject(s)
Dystroglycans/genetics , Muscular Dystrophies/congenital , Humans
16.
FEBS Lett ; 582(3): 439-44, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18201566

ABSTRACT

Alpha-dystroglycan (alpha-DG) plays a crucial role in maintaining the stability of muscle cell membrane. Although it has been shown that the N-terminal domain of alpha-DG (alpha-DG-N) is cleaved by a proprotein convertase, its physiological significance remains unclear. We show here that native alpha-DG-N is secreted by a wide variety of cultured cells into the culture media. The secreted alpha-DG-N was both N- and O-glycosylated. Finally, a small amount of alpha-DG-N was detectable in the normal human serum. These observations indicate that the cleavage of alpha-DG-N is a widespread event and suggest that the secreted alpha-DG-N might be transported via systemic circulation in vivo.


Subject(s)
Culture Media/metabolism , Dystroglycans/metabolism , Protein Processing, Post-Translational , Animals , Antibodies/chemistry , Antibodies/isolation & purification , COS Cells , Cells, Cultured , Chlorocebus aethiops , Culture Media/chemistry , Dystroglycans/chemistry , Dystroglycans/immunology , Glycosylation , HeLa Cells , Humans , Mice , Protein Structure, Tertiary , Serum/chemistry , Serum/metabolism
17.
J Neurochem ; 101(6): 1712-22, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17326765

ABSTRACT

Dystroglycan is a central component of the dystrophin-glycoprotein complex that links the extracellular matrix with cytoskeleton. Recently, mutations of the genes encoding putative glycosyltransferases were identified in several forms of congenital muscular dystrophies accompanied by brain anomalies and eye abnormalities, and aberrant glycosylation of alpha-dystroglycan has been implicated in their pathogeneses. These diseases are now collectively called alpha-dystroglycanopathy. In this study, we demonstrate that peripheral nerve myelination is defective in the fukutin-deficient chimeric mice, a mouse model of Fukuyama-type congenital muscular dystrophy, which is the most common alpha-dystroglycanopathy in Japan. In the peripheral nerve of these mice, the density of myelinated nerve fibers was significantly decreased and clusters of abnormally large non-myelinated axons were ensheathed by a single Schwann cell, indicating a defect of the radial sorting mechanism. The sugar chain moiety and laminin-binding activity of alpha-dystroglycan were severely reduced, while the expression of beta1-integrin was not altered in the peripheral nerve of the chimeric mice. We also show that the clustering of acetylcholine receptor is defective and neuromuscular junctions are fragmented in appearance in these mice. Expression of agrin and laminin as well as the binding activity of alpha-dystroglycan to these ligands was severely reduced at the neuromuscular junction. These results demonstrate that fukutin plays crucial roles in the myelination of peripheral nerve and formation of neuromuscular junction. They also suggest that defective glycosylation of alpha-dystroglycan may play a role in the impairment of these processes in the deficiency of fukutin.


Subject(s)
Chimera/genetics , Chimera/physiology , Muscular Dystrophy, Animal/physiopathology , Nerve Fibers, Myelinated/pathology , Neuromuscular Junction Diseases/genetics , Peripheral Nerves/physiopathology , Proteins/genetics , Agrin/metabolism , Animals , Dystroglycans/metabolism , Integrin beta1/metabolism , Laminin/metabolism , Mice , Neuromuscular Junction/ultrastructure , Peripheral Nervous System Diseases/pathology , Receptors, Cholinergic/metabolism , Transferases
18.
Biochem Biophys Res Commun ; 345(2): 867-71, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16701552

ABSTRACT

Dystroglycan (DG) complex, composed of alphaDG and betaDG, provides a link between the extracellular matrix (ECM) and cortical cytoskeleton. Although the proteolytic processing of betaDG was reported in various physiological and pathological conditions, its exact mechanism remains unknown. In this study, we addressed this issue using the cell culture system of rat schwannoma cell line RT4. We found that the culture medium of RT4 cells was enriched with the protease activity that degrades the fusion protein construct of the extracellular domain of betaDG specifically. This activity was suppressed by the inhibitor of matrix metalloproteinase-2 (MMP-2) and MMP-9, but not by the inhibitors of MMP-1, MMP-3, MMP-8, and MMP-13. Zymography and RT-PCR analysis showed that RT4 cells secreted MMP-2 and MMP-9 into the culture medium. Finally, active MMP-2 and MMP-9 enzymes degraded the fusion protein construct of the extracellular domain of betaDG. These results indicate (1) that RT4 cells secrete the protease activity that degrades the extracellular domain of betaDG specifically and (2) that MMP-2 and MMP-9 may be involved in this process.


Subject(s)
Dystroglycans/metabolism , Endopeptidases/metabolism , Extracellular Matrix/metabolism , Matrix Metalloproteinases/metabolism , Animals , Cells, Cultured , Collagenases/metabolism , Culture Media, Conditioned/metabolism , Cytoskeleton/metabolism , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13 , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
19.
J Stroke Cerebrovasc Dis ; 15(4): 176-7, 2006.
Article in English | MEDLINE | ID: mdl-17904072

ABSTRACT

Anterograde amnesia due to infarction of the anterior fornix is a rare but unique neuropsychological syndrome. Only 2 cases have been reported previously. Lacking focal neurologic deficits, this syndrome is not easy to diagnose. Moreover, it is not fully recognized by the clinicians, making its diagnosis all the more difficult. Here we report a patient of infarction of the anterior fornix and genu of the corpus callosum who developed sudden apathy and anterograde amnesia. Unfortunately, the patient was initially diagnosed and treated as an acute psychiatric disorder by a psychiatrist, and treatment for acute cerebral infarction could not be performed. This case emphasizes the importance of suspecting this syndrome and performing brain magnetic resonance imaging immediately in the patients presenting with anterograde amnesia of sudden onset.

20.
J Neurochem ; 95(2): 537-43, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16086681

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) family receptor alpha-1 (GFRalpha-1) is a receptor component of GDNF that associates with and activates the tyrosine kinase receptor Ret. To further understand GDNF and its receptor system in the PNS, we first characterized the expression of GFRalpha-1 in bovine peripheral nerve in vivo. GFRalpha-1 immunoreactivity was localized adjacent to the outermost layer of myelin sheath, as well as in the endoneurium and axoplasm. In a fractionation study, GFRalpha-1 was recovered mostly in the soluble fraction, although a small amount was recovered in the membrane fraction. A substantial amount of GFRalpha-1 in the membrane fraction was extractable by detergent and alkaline conditions. To further clarify the expression of GFRalpha-1 in Schwann cells, we examined cultured rat Schwann cells and the Schwannoma cell line RT4. Schwann cells expressed GFRalpha-1 in both the soluble/cytosolic and membrane fractions, and the membrane form of GFRalpha-1 was expressed at the outer surface of the Schwann cell plasma membrane. We also confirmed the secretion of the soluble form of GFRalpha-1 from Schwannoma cells in a metabolic labeling experiment. These data contribute to our knowledge of the production, expression and functions of GFRalpha-1 in the PNS.


Subject(s)
Peripheral Nerves/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Schwann Cells/metabolism , Animals , Biotin , Cattle , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Culture Media , Cytosol/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Immunoblotting , Immunohistochemistry , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurilemmoma/metabolism , Proto-Oncogene Proteins c-ret , Rats , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...