Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 16(11): 2752-2767, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34653404

ABSTRACT

Fukutin-related protein (FKRP) is a glycosyltransferase involved in glycosylation of alpha-dystroglycan (α-DG). Mutations in FKRP are associated with muscular dystrophies (MD) ranging from limb-girdle LGMDR9 to Walker-Warburg Syndrome (WWS), a severe type of congenital MD. Although hypoglycosylation of α-DG is the main hallmark of this group of diseases, a full understanding of the underlying pathophysiology is still missing. Here, we investigated molecular mechanisms impaired by FKRP mutations in pluripotent stem (PS) cell-derived myotubes. FKRP-deficient myotubes show transcriptome alterations in genes involved in extracellular matrix receptor interactions, calcium signaling, PI3K-Akt pathway, and lysosomal function. Accordingly, using a panel of patient-specific LGMDR9 and WWS induced PS cell-derived myotubes, we found a significant reduction in the autophagy-lysosome pathway for both disease phenotypes. In addition, we show that WWS myotubes display decreased ERK1/2 activity and increased apoptosis, which were restored in gene edited myotubes. Our results suggest the autophagy-lysosome pathway and apoptosis may contribute to the FKRP-associated MD pathogenesis.


Subject(s)
Apoptosis/genetics , Autophagy/genetics , Genetic Predisposition to Disease/genetics , Muscular Dystrophies/genetics , Mutation , Pentosyltransferases/genetics , Cell Line , Glycosylation , Humans , Lysosomes/genetics , Lysosomes/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Pentosyltransferases/metabolism , Pluripotent Stem Cells/metabolism , RNA-Seq/methods , Signal Transduction/genetics , Transcriptome/genetics , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
2.
Dis Model Mech ; 13(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32423971

ABSTRACT

The basal lamina is a specialized sheet of dense extracellular matrix (ECM) linked to the plasma membrane of specific cell types in their tissue context, which serves as a structural scaffold for organ genesis and maintenance. Disruption of the basal lamina and its functions is central to many disease processes, including cancer metastasis, kidney disease, eye disease, muscular dystrophies and specific types of brain malformation. The latter three pathologies occur in the α-dystroglycanopathies, which are caused by dysfunction of the ECM receptor α-dystroglycan. However, opportunities to study the basal lamina in various human disease tissues are restricted owing to its limited accessibility. Here, we report the generation of embryoid bodies from human induced pluripotent stem cells that model the basal lamina. Embryoid bodies cultured via this protocol mimic pre-gastrulation embryonic development, consisting of an epithelial core surrounded by a basal lamina and a peripheral layer of ECM-secreting endoderm. In α-dystroglycanopathy patient embryoid bodies, electron and fluorescence microscopy reveal ultrastructural basal lamina defects and reduced ECM accumulation. By starting from patient-derived cells, these results establish a method for the in vitro synthesis of patient-specific basal lamina and recapitulate disease-relevant ECM defects seen in the α-dystroglycanopathies. Finally, we apply this system to evaluate an experimental ribitol supplement therapy on genetically diverse α-dystroglycanopathy patient samples.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Basement Membrane/metabolism , Embryoid Bodies/metabolism , Extracellular Matrix/metabolism , Human Embryonic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Walker-Warburg Syndrome/metabolism , Basement Membrane/drug effects , Basement Membrane/ultrastructure , Case-Control Studies , Cell Culture Techniques , Cells, Cultured , Child , Child, Preschool , Dystroglycans/genetics , Dystroglycans/metabolism , Embryoid Bodies/drug effects , Embryoid Bodies/ultrastructure , Extracellular Matrix/drug effects , Extracellular Matrix/ultrastructure , Female , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/ultrastructure , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/ultrastructure , Infant, Newborn , Male , Middle Aged , Ribitol/pharmacology , Walker-Warburg Syndrome/drug therapy , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/pathology
3.
Mol Ther ; 28(2): 664-676, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31843448

ABSTRACT

Patients with α-dystroglycanopathies, a subgroup of rare congenital muscular dystrophies, present with a spectrum of clinical manifestations that includes muscular dystrophy as well as CNS and ocular abnormalities. Although patients with α-dystroglycanopathies are genetically heterogeneous, they share a common defect of aberrant post-translational glycosylation modification of the dystroglycan alpha-subunit, which renders it defective in binding to several extracellular ligands such as laminin-211 in skeletal muscles, agrin in neuromuscular junctions, neurexin in the CNS, and pikachurin in the eye, leading to various symptoms. The genetic heterogeneity associated with the development of α-dystroglycanopathies poses significant challenges to developing a generalized treatment to address the spectrum of genetic defects. Here, we propose the development of a bispecific antibody (biAb) that functions as a surrogate molecular linker to reconnect laminin-211 and the dystroglycan beta-subunit to ameliorate sarcolemmal fragility, a primary pathology in patients with α-dystroglycan-related muscular dystrophies. We show that the treatment of LARGEmyd-3J mice, an α-dystroglycanopathy model, with the biAb improved muscle function and protected muscles from exercise-induced damage. These results demonstrate the viability of a biAb that binds to laminin-211 and dystroglycan simultaneously as a potential treatment for patients with α-dystroglycanopathy.


Subject(s)
Antibodies, Bispecific/pharmacology , Dystroglycans/metabolism , Laminin/metabolism , Walker-Warburg Syndrome/metabolism , Animals , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Disease Models, Animal , Dystroglycans/immunology , Gene Expression , Humans , Immunohistochemistry , Injections, Intramuscular , Laminin/genetics , Laminin/immunology , Mice , Mice, Knockout , Models, Biological , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Protein Binding/drug effects , Protein Interaction Domains and Motifs/genetics , Sarcolemma/drug effects , Sarcolemma/metabolism , Walker-Warburg Syndrome/drug therapy , Walker-Warburg Syndrome/etiology
4.
Hum Mol Genet ; 28(20): 3369-3390, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31348492

ABSTRACT

Congenital muscular dystrophy type-1A (Lama2-CMD) and Duchenne muscular dystrophy (DMD) result from deficiencies of laminin-α2 and dystrophin proteins, respectively. Although both proteins strengthen the sarcolemma, they are implicated in clinically distinct phenotypes. We used RNA-deep sequencing (RNA-Seq) of dy2J/dy2J, Lama2-CMD mouse model, skeletal muscle at 8 weeks of age to elucidate disease pathophysiology. This study is the first report of dy2J/dy2J model whole transcriptome profile. RNA-Seq of the mdx mouse model of DMD and wild-type (WT) mouse was carried as well in order to enable a novel comparison of dy2J/dy2J to mdx. A large group of shared differentially expressed genes (DEGs) was found in dy2J/dy2J and mdx models (1834 common DEGs, false discovery rate [FDR] < 0.05). Enrichment pathway analysis using ingenuity pathway analysis showed enrichment of inflammation, fibrosis, cellular movement, migration and proliferation of cells, apoptosis and necrosis in both mouse models (P-values 3E-10-9E-37). Via canonical pathway analysis, actin cytoskeleton, integrin, integrin-linked kinase, NF-kB, renin-angiotensin, epithelial-mesenchymal transition, and calcium signaling were also enriched and upregulated in both models (FDR < 0.05). Interestingly, significant downregulation of Pax7 was detected in dy2J/dy2J compared to upregulation of this key regeneration gene in mdx mice. Pax3 and Mamstr genes were also downregulated in dy2J/dy2J compared to WT mice. These results may explain the distinct disease course and severity in these models. While the mdx model at that stage shows massive regeneration, the dy2J/dy2J shows progressive dystrophic process. Our data deepen our understanding of the molecular pathophysiology and suggest new targets for additional therapies to upregulate regeneration in Lama2-CMD.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , PAX3 Transcription Factor/metabolism , PAX7 Transcription Factor/metabolism , Animals , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Disease Models, Animal , Male , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/genetics , Necrosis/genetics , Necrosis/metabolism , PAX3 Transcription Factor/genetics , PAX7 Transcription Factor/genetics , Real-Time Polymerase Chain Reaction , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism
5.
Sci Rep ; 8(1): 8543, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867208

ABSTRACT

Hypoglycosylation of α-dystroglycan (α-DG) resulting from deficiency of protein O-mannosyltransferase 1 (POMT1) may cause severe neuromuscular dystrophies with brain and eye anomalies, named dystroglycanopathies. The retinal involvement of these disorders motivated us to generate a conditional knockout (cKO) mouse experiencing a Pomt1 intragenic deletion (exons 3-4) during the development of photoreceptors, mediated by the Cre recombinase expressed from the cone-rod homeobox (Crx) gene promoter. In this mouse, retinal α-DG was unglycosylated and incapable of binding laminin. Retinal POMT1 deficiency caused significant impairments in both electroretinographic recordings and optokinetic reflex in Pomt1 cKO mice, and immunohistochemical analyses revealed the absence of ß-DG and of the α-DG-interacting protein, pikachurin, in the outer plexiform layer (OPL). At the ultrastructural level, noticeable alterations were observed in the ribbon synapses established between photoreceptors and bipolar cells. Therefore, O-mannosylation of α-DG in the retina carried out by POMT1 is crucial for the establishment of proper synapses at the OPL and transmission of visual information from cones and rods to their postsynaptic neurons.


Subject(s)
Electroretinography , Mannosyltransferases , Retinal Cone Photoreceptor Cells , Synapses , Walker-Warburg Syndrome , Animals , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Mannosyltransferases/genetics , Mannosyltransferases/metabolism , Mice , Mice, Knockout , Retinal Cone Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Synapses/genetics , Synapses/metabolism , Synapses/pathology , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
6.
Mol Vis ; 24: 43-58, 2018.
Article in English | MEDLINE | ID: mdl-29416295

ABSTRACT

Purpose: Dystroglycanopathies are a heterogeneous group of recessive neuromuscular dystrophies that affect the muscle, brain and retina, and are caused by deficiencies in the O-glycosylation of α-dystroglycan. This post-translational modification is essential for the formation and maintenance of ribbon synapses in the retina. Fukutin and fukutin-related protein (FKRP) are two glycosyltransferases whose deficiency is associated with severe dystroglycanopathies. These enzymes carry out in vitro the addition of a tandem ribitol 5-phosphate moiety to the so-called core M3 phosphotrisaccharide of α-dystroglycan. However, their expression pattern and function in the healthy mammalian retina has not so far been investigated. In this work, we have addressed the expression of the FKTN (fukutin) and FKRP genes in the retina of mammals, and characterized the distribution pattern of their protein products in the adult mouse retina and the 661W photoreceptor cell line. Methods: By means of reverse transcription (RT)-PCR and immunoblotting, we have studied the expression at the mRNA and protein levels of the fukutin and FKRP genes in different mammalian species, from rodents to humans. Immunofluorescence confocal microscopy analyses were performed to characterize the distribution profile of their protein products in mouse retinal sections and in 661W cultured cells. Results: Both genes were expressed at the mRNA and protein levels in the neural retina of all mammals studied. Fukutin was present in the cytoplasmic and nuclear fractions in the mouse retina and 661W cells, and accumulated in the endoplasmic reticulum. FKRP was located in the cytoplasmic fraction in the mouse retina and concentrated in the Golgi complex. However, and in contrast to retinal tissue, FKRP additionally accumulated in the nucleus of the 661W photoreceptors. Conclusions: Our results suggest that fukutin and FKRP not only participate in the synthesis of O-mannosyl glycans added to α-dystroglycan in the endoplasmic reticulum and Golgi complex, but that they could also play a role, that remains to be established, in the nucleus of retinal neurons.


Subject(s)
Dystroglycans/genetics , Membrane Proteins/genetics , Protein Processing, Post-Translational , Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Animals , Cattle , Cell Line , Dystroglycans/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Gene Expression , Genes, Recessive , Glycosylation , Golgi Apparatus/genetics , Golgi Apparatus/metabolism , Humans , Macaca fascicularis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Pentosyltransferases , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Retinal Cone Photoreceptor Cells/cytology , Signal Transduction , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
7.
Biochim Biophys Acta Gen Subj ; 1861(10): 2462-2472, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28711406

ABSTRACT

BACKGROUND: O-mannosyl glycans have been found in a limited number of glycoproteins of the brain, nerves, and skeletal muscles, particularly in α-dystroglycan (α-DG). Defects in O-mannosyl glycan on α-DG are the primary cause of a group of congenital muscular dystrophies, which are collectively termed α-dystroglycanopathy. Recent studies have revealed various O-mannosyl glycan structures, which can be classified as core M1, core M2, and core M3 glycans. Although many dystroglycanopathy genes are involved in core M3 processing, the structure and biosynthesis of core M3 glycan remains only partially understood. SCOPE OF REVIEW: This review presents recent findings about the structure, biosynthesis, and pathology of O-mannosyl glycans. MAJOR CONCLUSIONS: Recent studies have revealed that the entire structure of core M3 glycan, including ribitol-5-phosphate, is a novel structure in mammals; its unique biosynthetic pathway has been elucidated by the identification of new causative genes for α-dystroglycanopathies and their functions. GENERAL SIGNIFICANCE: O-mannosyl glycan has a novel, unique structure that is important for the maintenance of brain and muscle functions. These findings have opened up a new field in glycoscience. These studies will further contribute to the understanding of the pathomechanism of α-dystroglycanopathy and the development of glycotherapeutics. This article is part of a Special Issue entitled Neuro-glycoscience, edited by Kenji Kadomatsu and Hiroshi Kitagawa.


Subject(s)
Dystroglycans/chemistry , Muscular Dystrophies/metabolism , N-Acetylglucosaminyltransferases/chemistry , Pentosephosphates/metabolism , Protein Processing, Post-Translational , Walker-Warburg Syndrome/metabolism , Animals , Brain/metabolism , Brain/pathology , Carbohydrate Sequence , Dystroglycans/genetics , Dystroglycans/metabolism , Glycosylation , Humans , Mannose/chemistry , Mannose/metabolism , Models, Molecular , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Pentosephosphates/chemistry , Polysaccharides/chemistry , Polysaccharides/metabolism , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/pathology
8.
Mol Aspects Med ; 51: 115-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27421908

ABSTRACT

α-Dystroglycanopathy, an autosomal recessive disease, is associated with the development of a variety of diseases, including muscular dystrophy. In humans, α-dystroglycanopathy includes various types of congenital muscular dystrophy such as Fukuyama type congenital muscular dystrophy (FCMD), muscle eye brain disease (MEB), and the Walker Warburg syndrome (WWS), and types of limb girdle muscular dystrophy 2I (LGMD2I). α-Dystroglycanopathy share a common etiology, since it is invariably caused by gene mutations that are associated with the O-mannose glycosylation pathway of α-dystroglycan (α-DG). α-DG is a central member of the dystrophin glycoprotein complex (DGC) family in peripheral membranes, and the proper glycosylation of α-DG is essential for it to bind to extracellular matrix proteins, such as laminin, to cell components. The disruption of this ligand-binding is thought to result in damage to cell membrane integration, leading to the development of muscular dystrophy. Clinical manifestations of α-dystroglycanopathy frequently include mild to severe alterations in the central nervous system and optical manifestations in addition to muscular dystrophy. Eighteen causative genes for α-dystroglycanopathy have been identified to date, and it is likely that more will be reported in the near future. These findings have stimulated extensive and energetic investigations in this research field, and novel glycosylation pathways have been implicated in the process. At the same time, the use of gene therapy, antisense therapy, and enzymatic supplementation have been evaluated as therapeutic possibilities for some types of α-dystroglycanopathy. Here we review the molecular and clinical findings associated with α-dystroglycanopathy and the development of therapeutic approaches, by comparing the approaches with the development of Duchenne muscular dystrophy.


Subject(s)
Walker-Warburg Syndrome , Biomedical Research , Dystroglycans/chemistry , Dystroglycans/metabolism , Enzyme Replacement Therapy , Genetic Therapy , Glycosyltransferases , Humans , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/therapy
9.
Trends Mol Med ; 22(7): 615-628, 2016 07.
Article in English | MEDLINE | ID: mdl-27263464

ABSTRACT

A key aspect of cellular function is the proper assembly and utilization of ribonucleoproteins (RNPs). Recent studies have shown that hyper- or hypo-assembly of various RNPs can lead to human diseases. Defects in the formation of RNPs lead to 'RNP hypo-assembly diseases', which can be caused by RNA degradation outcompeting RNP assembly. By contrast, excess RNP assembly, either in higher order RNP granules, or due to the expression of repeat-containing RNAs, can lead to 'RNP hyper-assembly diseases'. Here, we discuss the most recent advances in understanding the cause of disease onset, as well as potential therapies from the aspect of modulating RNP assembly in the cell, which presents a novel route to the treatment of these diseases.


Subject(s)
Dyskeratosis Congenita/metabolism , Muscular Atrophy, Spinal/metabolism , Ribonucleoproteins/metabolism , Animals , Dwarfism/genetics , Dwarfism/metabolism , Dwarfism/pathology , Dyskeratosis Congenita/genetics , Dyskeratosis Congenita/pathology , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Hair/abnormalities , Hair/metabolism , Hair/pathology , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Hirschsprung Disease/pathology , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Immunologic Deficiency Syndromes/pathology , Mallory Bodies/genetics , Mallory Bodies/metabolism , Mallory Bodies/pathology , Microcephaly/genetics , Microcephaly/metabolism , Microcephaly/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Mutation , Osteochondrodysplasias/congenital , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Primary Immunodeficiency Diseases , RNA Stability , Ribonucleoproteins/analysis , Ribonucleoproteins/genetics , Scoliosis/genetics , Scoliosis/metabolism , Scoliosis/pathology , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
10.
Skelet Muscle ; 6: 3, 2016.
Article in English | MEDLINE | ID: mdl-26900448

ABSTRACT

BACKGROUND: The defective glycosylation of α-dystroglycan is associated with a group of muscular dystrophies that are collectively referred to as the secondary dystroglycanopathies. Mutations in the gene encoding fukutin-related protein (FKRP) are one of the most common causes of secondary dystroglycanopathy in the UK and are associated with a wide spectrum of disease. Whilst central nervous system involvement has a prenatal onset, no studies have addressed prenatal muscle development in any of the mouse models for this group of diseases. In view of the pivotal role of α-dystroglycan in early basement membrane formation, we sought to determine if the muscle formation was altered in a mouse model of FKRP-related dystrophy. RESULTS: Mice with a knock-down in FKRP (FKRP(KD)) showed a marked reduction in α-dystroglycan glycosylation and reduction in laminin binding by embryonic day 15.5 (E15.5), relative to wild type controls. In addition, the total number of Pax7(+) progenitor cells in the FKRP(KD) tibialis anterior at E15.5 was significantly reduced, and myotube cluster/myofibre size showed a significant reduction in size. Moreover, myoblasts isolated from the limb muscle of these mice at E15.5 showed a marked reduction in their ability to form myotubes in vitro. CONCLUSIONS: These data identify an early reduction of laminin α2, reduction of myogenicity and depletion of Pax7(+) progenitor cells which would be expected to compromise subsequent postnatal muscle growth and its ability to regenerate postnatally. These findings are of significance to the development of future therapies in this group of devastating conditions.


Subject(s)
Muscle Development , Muscle, Skeletal/physiopathology , Walker-Warburg Syndrome/physiopathology , Animals , Cells, Cultured , Disease Models, Animal , Dystroglycans/metabolism , Genetic Predisposition to Disease , Gestational Age , Glycosylation , Laminin/metabolism , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , PAX7 Transcription Factor/metabolism , Pentosyltransferases , Phenotype , Protein Processing, Post-Translational , Proteins/genetics , Proteins/metabolism , Transferases , Walker-Warburg Syndrome/embryology , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism
11.
Sci Rep ; 5: 8316, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25661440

ABSTRACT

α-Dystroglycanopathy (α-DGP) is a group of muscular dystrophy characterized by abnormal glycosylation of α-dystroglycan (α-DG), including Fukuyama congenital muscular dystrophy (FCMD), muscle-eye-brain disease, Walker-Warburg syndrome, and congenital muscular dystrophy type 1D (MDC1D), etc. LARGE, the causative gene for MDC1D, encodes a glycosyltransferase to form [-3Xyl-α1,3GlcAß1-] polymer in the terminal end of the post-phosphoryl moiety, which is essential for α-DG function. It has been proposed that LARGE possesses the great potential to rescue glycosylation defects in α-DGPs regardless of causative genes. However, the in vivo therapeutic benefit of using LARGE activity is controversial. To explore the conditions needed for successful LARGE gene therapy, here we used Large-deficient and fukutin-deficient mouse models for MDC1D and FCMD, respectively. Myofibre-selective LARGE expression via systemic adeno-associated viral gene transfer ameliorated dystrophic pathology of Large-deficient mice even when intervention occurred after disease manifestation. However, the same strategy failed to ameliorate the dystrophic phenotype of fukutin-conditional knockout mice. Furthermore, forced expression of Large in fukutin-deficient embryonic stem cells also failed to recover α-DG glycosylation, however coexpression with fukutin strongly enhanced α-DG glycosylation. Together, our data demonstrated that fukutin is required for LARGE-dependent rescue of α-DG glycosylation, and thus suggesting new directions for LARGE-utilizing therapy targeted to myofibres.


Subject(s)
Gene Expression , Muscle Fibers, Skeletal/metabolism , N-Acetylglucosaminyltransferases/genetics , Proteins/genetics , Walker-Warburg Syndrome/genetics , Animals , Dependovirus/genetics , Disease Models, Animal , Dystroglycans/metabolism , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Glycosylation , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Proteins/metabolism , Transduction, Genetic , Transferases , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/therapy
12.
Hum Mol Genet ; 24(8): 2241-6, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25552652

ABSTRACT

Binding of cellular α-dystroglycan (α-DG) to its extracellular matrix ligands is fully dependent on a unique O-mannose-linked glycan. Disrupted O-mannosylation is the hallmark of the muscular dystrophy-dystroglycanopathy (MDDG) syndromes. SLC35A1, encoding the transporter of cytidine 5'-monophosphate-sialic acid, was recently identified as MDDG candidate gene. This is surprising, since sialic acid itself is dispensable for α-DG-ligand binding. In a novel SLC35A1-deficient cell model, we demonstrated a lack of α-DG O-mannosylation, ligand binding and incorporation of sialic acids. Removal of sialic acids from HAP1 wild-type cells after incorporation or preventing sialylation during synthesis did not affect α-DG O-mannosylation or ligand binding but did affect sialylation. Lentiviral-mediated complementation with the only known disease mutation p.Q101H failed to restore deficient O-mannosylation in SLC35A1 knockout cells and partly restored sialylation. These data indicate a role for SLC35A1 in α-DG O-mannosylation that is distinct from sialic acid metabolism. In addition, human SLC35A1 deficiency can be considered as a combined disorder of α-DG O-mannosylation and sialylation, a novel variant of the MDDG syndromes.


Subject(s)
Dystroglycans/metabolism , Mannose/metabolism , Nucleotide Transport Proteins/genetics , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism , Cell Line , Cytidine Monophosphate/metabolism , Humans , Mutation , N-Acetylneuraminic Acid/metabolism , Nucleotide Transport Proteins/metabolism
13.
PLoS One ; 9(9): e106721, 2014.
Article in English | MEDLINE | ID: mdl-25198651

ABSTRACT

Defects in dystroglycan glycosylation are associated with a group of muscular dystrophies, termed dystroglycanopathies, that include Fukuyama congenital muscular dystrophy (FCMD). It is widely believed that abnormal glycosylation of dystroglycan leads to disease-causing membrane fragility. We previously generated knock-in mice carrying a founder retrotransposal insertion in fukutin, the gene responsible for FCMD, but these mice did not develop muscular dystrophy, which hindered exploring therapeutic strategies. We hypothesized that dysferlin functions may contribute to muscle cell viability in the knock-in mice; however, pathological interactions between glycosylation abnormalities and dysferlin defects remain unexplored. To investigate contributions of dysferlin deficiency to the pathology of dystroglycanopathy, we have crossed dysferlin-deficient dysferlin(sjl/sjl) mice to the fukutin-knock-in fukutin(Hp/-) and Large-deficient Largemyd/myd mice, which are phenotypically distinct models of dystroglycanopathy. The fukutin(Hp/-) mice do not show a dystrophic phenotype; however, (dysferlin(sjl/sjl): fukutin(Hp/-)) mice showed a deteriorated phenotype compared with (dysferlinsjl/sjl: fukutin(Hp/+)) mice. These data indicate that the absence of functional dysferlin in the asymptomatic fukutin(Hp/-) mice triggers disease manifestation and aggravates the dystrophic phenotype. A series of pathological analyses using double mutant mice for Large and dysferlin indicate that the protective effects of dysferlin appear diminished when the dystrophic pathology is severe and also may depend on the amount of dysferlin proteins. Together, our results show that dysferlin exerts protective effects on the fukutin(Hp/-) FCMD mouse model, and the (dysferlin(sjl/sjl): fukutin(Hp/-)) mice will be useful as a novel model for a recently proposed antisense oligonucleotide therapy for FCMD.


Subject(s)
Dystroglycans/metabolism , Membrane Proteins/genetics , Models, Biological , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Walker-Warburg Syndrome/genetics , Animals , Dysferlin , Humans , Mice , Mice, Transgenic , Walker-Warburg Syndrome/metabolism
14.
Brain Dev ; 36(8): 730-3, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24113355

ABSTRACT

We report the first case of Leigh syndrome (LS) with Fukuyama congenital muscular dystrophy (FCMD). A neonate suffered from lactic acidosis and subsequently presented with poor feeding, muscle weakness, hypotonia, cardiopulmonary dysfunction, and hydrocephalus. He died at 17 months. The findings of brain magnetic resonance imaging indicated some specific features of both LS and FCMD, and FCMD gene mutation was detected. Decreased mitochondrial respiratory complex I and II activity was noted. Mitochondrial DNA sequencing showed no pathogenic mutation. A case with complex I+II deficiency has rarely been reported, suggesting a nuclear gene mutation.


Subject(s)
Leigh Disease/complications , Leigh Disease/diagnosis , Walker-Warburg Syndrome/complications , Walker-Warburg Syndrome/diagnosis , Brain/pathology , Humans , Infant, Newborn , Leigh Disease/pathology , Magnetic Resonance Imaging , Male , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
15.
Nat Commun ; 4: 2935, 2013.
Article in English | MEDLINE | ID: mdl-24343302

ABSTRACT

Tumour cells primarily utilize aerobic glycolysis for energy production, a phenomenon known as the Warburg effect. Its mechanism is not well understood. The tumour suppressor gene p53 is frequently mutated in tumours. Many tumour-associated mutant p53 (mutp53) proteins not only lose tumour suppressive function but also gain new oncogenic functions that are independent of wild-type p53, defined as mutp53 gain of function (GOF). Here we show that tumour-associated mutp53 stimulates the Warburg effect in cultured cells and mutp53 knockin mice as a new mutp53 GOF. Mutp53 stimulates the Warburg effect through promoting GLUT1 translocation to the plasma membrane, which is mediated by activated RhoA and its downstream effector ROCK. Inhibition of RhoA/ROCK/GLUT1 signalling largely abolishes mutp53 GOF in stimulating the Warburg effect. Furthermore, inhibition of glycolysis in tumour cells greatly compromises mutp53 GOF in promoting tumorigenesis. Thus, our results reveal a new mutp53 GOF and a mechanism for controlling the Warburg effect.


Subject(s)
Genes, p53 , Mutation , Neoplasms/genetics , Walker-Warburg Syndrome/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cells, Cultured , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Humans , Lactates/blood , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Walker-Warburg Syndrome/genetics , Xenograft Model Antitumor Assays , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
16.
No To Hattatsu ; 45(6): 436-9, 2013 Nov.
Article in Japanese | MEDLINE | ID: mdl-24313002

ABSTRACT

OBJECTIVE: Sudden unexpected death (SUD) may occur in patients with Fukuyama congenital muscular dystrophy (FCMD). In this study, we performed immunohistochemical examination of SUD-related functional markers in the brainstem of autopsy cases of FCMD, in order to clarify the pathogenesis of SUD. METHODS: The examination was conducted on 9 autopsy cases of FCMD, including a case of SUD and 3 of acute death (AD) in which SUD was suspected but not confirmed. We immunohistochemically examined serial brainstem sections for serotonin and catecholamine neurons, neuropeptides, and c-Fos, a neuron activation marker. RESULTS: 1) Number of serotonin neurons was reduced in 7 cases, including the cases of SUD and AD. 2) Expressions of neuropeptides were exaggerated in the spinal trigeminal nucleus in 5 cases, including the SUD and AD ones. 3) Neurons immunoreactive for c-Fos were found in 3 cases, including the SUD and AD cases. 4) The suspected case of SUD showed changes in all SUD markers. CONCLUSIONS: Changes in the tested markers were found predominantly in the SUD and AD cases, indicating functional fragility in the brainstem of patients with FCMD.


Subject(s)
Brain Stem/metabolism , Death, Sudden/pathology , Neurons/metabolism , Walker-Warburg Syndrome/metabolism , Adolescent , Adult , Autopsy/methods , Brain Stem/pathology , Child , Female , Humans , Immunohistochemistry/methods , Male , Walker-Warburg Syndrome/pathology , Young Adult
17.
Biochem Biophys Res Commun ; 440(1): 88-93, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-24041696

ABSTRACT

Hypoglycosylation is a common characteristic of dystroglycanopathy, which is a group of congenital muscular dystrophies. More than ten genes have been implicated in α-dystroglycanopathies that are associated with the defect in the O-mannosylation pathway. One such gene is GTDC2, which was recently reported to encode O-mannose ß-1,4-N-acetylglucosaminyltransferase. Here we show that GTDC2 generates CTD110.6 antibody-reactive N-acetylglucosamine (GlcNAc) epitopes on the O-mannosylated α-dystroglycan (α-DG). Using the antibody, we show that mutations of GTDC2 identified in Walker-Warburg syndrome and alanine-substitution of conserved residues between GTDC2 and EGF domain O-GlcNAc transferase resulted in decreased glycosylation. Moreover, GTDC2-modified GlcNAc epitopes are localized in the endoplasmic reticulum (ER). These data suggested that GTDC2 is a novel glycosyltransferase catalyzing GlcNAcylation of O-mannosylated α-DG in the ER. CTD110.6 antibody may be useful to detect a specific form of GlcNAcylated O-mannose and to analyze defective O-glycosylation in α-dystroglycanopathies.


Subject(s)
Acetylglucosamine/metabolism , Dystroglycans/metabolism , Endoplasmic Reticulum/metabolism , Epitopes/metabolism , Glycosyltransferases/metabolism , Acetylglucosamine/immunology , Animals , Antibodies/immunology , Dystroglycans/chemistry , Dystroglycans/immunology , Endoplasmic Reticulum/immunology , Epitopes/immunology , Glycosylation , Glycosyltransferases/genetics , Glycosyltransferases/immunology , HEK293 Cells , Humans , Mutation , Protein Structure, Tertiary , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/immunology , Walker-Warburg Syndrome/metabolism
18.
Int J Cancer ; 133(10): 2504-10, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23661584

ABSTRACT

The reprogramming of cellular metabolism in cancer cells is a well-documented effect. It has previously been shown that common oncogene expression can induce aerobic glycolysis in cancer cells. However, the direct effect of an inflammatory microenvironment on cancer cell metabolism is not known. Here, we illustrate that treatment of nonmalignant (MCF-10a) and malignant (MCF-7) breast epithelial cells with low-level (10 ng/ml) tumor necrosis factor alpha (TNF-α) significantly increased glycolytic reliance, lactate export and expression of the glucose transporter 1 (GLUT1). TNF-α decreased total mitochondrial content; however, oxygen consumption rate was not significantly altered, suggesting that overall mitochondrial function was increased. Upon glucose starvation, MCF7 cells treated with TNF-α demonstrated significantly lower viability than nontreated cells. Interestingly, these properties can be partially reversed by coincubation with the anti-inflammatory agent curcumin in a dose-dependent manner. This work demonstrates that aerobic glycolysis can be directly induced by an inflammatory microenvironment independent of additional genetic mutations and signals from adjacent cells. Furthermore, we have identified that a natural dietary compound can reverse this effect.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Breast/drug effects , Curcumin/pharmacology , Epithelial Cells/drug effects , Tumor Necrosis Factor-alpha/metabolism , Walker-Warburg Syndrome/metabolism , Breast/cytology , Breast/metabolism , Cell Line, Tumor , Epithelial Cells/metabolism , Glucose/metabolism , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lactic Acid/metabolism , MCF-7 Cells , Mitochondria/pathology , NF-kappa B/metabolism , Oxygen Consumption/drug effects , Tumor Microenvironment/drug effects
19.
Am J Hum Genet ; 92(3): 468-74, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23472759

ABSTRACT

Cobblestone brain malformation (COB) is a neuronal migration disorder characterized by protrusions of neurons beyond the first cortical layer at the pial surface of the brain. It is usually seen in association with dystroglycanopathy types of congenital muscular dystrophies (CMDs) and ocular abnormalities termed muscle-eye-brain disease. Here we report homozygous deleterious mutations in LAMB1, encoding laminin subunit beta-1, in two families with autosomal-recessive COB. Affected individuals displayed a constellation of brain malformations including cortical gyral and white-matter signal abnormalities, severe cerebellar dysplasia, brainstem hypoplasia, and occipital encephalocele, but they had less apparent ocular or muscular abnormalities than are typically observed in COB. LAMB1 is localized to the pial basement membrane, suggesting that defective connection between radial glial cells and the pial surface mediated by LAMB1 leads to this malformation.


Subject(s)
Brain/abnormalities , Laminin/genetics , Muscular Dystrophies/genetics , Nervous System Malformations/genetics , Sequence Deletion , Walker-Warburg Syndrome/genetics , Basement Membrane/metabolism , Basement Membrane/pathology , Brain/metabolism , Brain/pathology , Cerebellum/metabolism , Cerebellum/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Encephalocele/genetics , Encephalocele/metabolism , Encephalocele/pathology , Female , Genetic Predisposition to Disease , Homozygote , Humans , Male , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Nervous System Malformations/metabolism , Nervous System Malformations/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Walker-Warburg Syndrome/metabolism , Walker-Warburg Syndrome/pathology
20.
Biochem Biophys Res Commun ; 424(2): 354-7, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22771323

ABSTRACT

Mutations in the gene encoding fukutin protein cause Fukuyama muscular dystrophy, a severe congenital disorder that occurs mainly in Japan. A major consequence of the mutation is reduced glycosylation of alpha-dystroglycan, which is also a feature of other forms of congenital and limb-girdle muscular dystrophy. Immunodetection of endogenous fukutin in cells and tissues has been difficult and this has hampered progress in understanding fukutin function and disease pathogenesis. Using a new panel of monoclonal antibodies which bind to different defined sites on the fukutin molecule, we now show that fukutin has the predicted size for a protein without extensive glycosylation and is present at the Golgi apparatus at very low levels. These antibodies should enable more rapid future progress in understanding the molecular function of fukutin.


Subject(s)
Antibodies, Monoclonal , Membrane Proteins/analysis , Walker-Warburg Syndrome/diagnosis , Amino Acid Sequence , Animals , Epitope Mapping , Glycosylation , Golgi Apparatus/metabolism , HeLa Cells , Humans , Hybridomas , Immunodominant Epitopes/analysis , Immunodominant Epitopes/genetics , Immunodominant Epitopes/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Library , Walker-Warburg Syndrome/genetics , Walker-Warburg Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...