Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Publication year range
1.
Exp Cell Res ; 440(1): 114118, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38852763

ABSTRACT

Autophagy phenomenon in the cell maintains proteostasis balance by eliminating damaged organelles and protein aggregates. Imbalance in autophagic flux may cause accumulation of protein aggregates in various neurodegenerative disorders. Regulation of autophagy by either calcium or chaperone play a key role in the removal of protein aggregates from the cell. The neuromuscular rare genetic disorder, GNE Myopathy, is characterized by accumulation of rimmed vacuoles having protein aggregates of ß-amyloid and tau that may result from altered autophagic flux. In the present study, the autophagic flux was deciphered in HEK cell-based model for GNE Myopathy harbouring GNE mutations of Indian origin. The refolding activity of HSP70 chaperone was found to be reduced in GNE mutant cells compared to wild type controls. The autophagic markers LC3II/I ratio was altered with increased number of autophagosome formation in GNE mutant cells compared to wild type cells. The cytosolic calcium levels were also increased in GNE mutant cells of Indian origin. Interestingly, treatment of GNE mutant cells with HSP70 activator, BGP-15, restored the expression and refolding activity of HSP70 along with autophagosome formation. Treatment with calcium chelator, BAPTA-AM restored the cytoplasmic calcium levels and autophagosome formation but not LC3II/I ratio significantly. Our study provides insights towards GNE mutation specific response for autophagy regulation and opens up a therapeutic advancement area in calcium signalling and HSP70 function for GNE related Myopathy.


Subject(s)
Autophagy , Calcium , Distal Myopathies , HSP70 Heat-Shock Proteins , Multienzyme Complexes , Mutation , Humans , Autophagy/genetics , Autophagy/drug effects , Mutation/genetics , Calcium/metabolism , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , HEK293 Cells , Autophagosomes/metabolism , Autophagosomes/drug effects , India
2.
Proc Natl Acad Sci U S A ; 119(38): e2122523119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36112647

ABSTRACT

T cell intracellular antigen-1 (TIA-1) plays a central role in stress granule (SG) formation by self-assembly via the prion-like domain (PLD). In the TIA-1 PLD, amino acid mutations associated with neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) or Welander distal myopathy (WDM), have been identified. However, how these mutations affect PLD self-assembly properties has remained elusive. In this study, we uncovered the implicit pathogenic structures caused by the mutations. NMR analysis indicated that the dynamic structures of the PLD are synergistically determined by the physicochemical properties of amino acids in units of five residues. Molecular dynamics simulations and three-dimensional electron crystallography, together with biochemical assays, revealed that the WDM mutation E384K attenuated the sticky properties, whereas the ALS mutations P362L and A381T enhanced the self-assembly by inducing ß-sheet interactions and highly condensed assembly, respectively. These results suggest that the P362L and A381T mutations increase the likelihood of irreversible amyloid fibrillization after phase-separated droplet formation, and this process may lead to pathogenicity.


Subject(s)
Amino Acids , Amyotrophic Lateral Sclerosis , Prions , Protein Aggregation, Pathological , T-Cell Intracellular Antigen-1 , Amino Acids/chemistry , Amino Acids/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Distal Myopathies/genetics , Distal Myopathies/metabolism , Humans , Mutation , Prions/chemistry , Protein Aggregation, Pathological/genetics , Protein Conformation, beta-Strand/genetics , Protein Domains/genetics , T-Cell Intracellular Antigen-1/chemistry , T-Cell Intracellular Antigen-1/genetics
3.
J Muscle Res Cell Motil ; 43(2): 49-61, 2022 06.
Article in English | MEDLINE | ID: mdl-35524895

ABSTRACT

Mutations in the sialic acid biosynthesis enzyme GNE lead to a late-onset, debilitating neuromuscular disorder, GNE myopathy, characterized by progressive skeletal muscle weakness. The mechanisms responsible for skeletal muscle specificity, late-onset, and disease progression are unknown. Our main aim is to understand the reason for skeletal muscle-specific phenotype. To answer this question, we have analyzed the expression profile of the GNE gene and its multiple mRNA variants in different human tissues. A combinatorial approach encompassing bioinformatics tools and molecular biology techniques was used. NCBI, Ensembl, and GTEx were used for data mining. The expression analysis of GNE and its variants was performed with cDNA tissue panel using PCR and targeted RNA-seq. Among nine different GNE isoforms reported in this study, transcript variants 1, X1, and X2 were not tissue specific. Transcript variants 1, 6, X1, and X2, were found in skeletal muscles suggesting their possible role in GNE myopathy. In the current study, we present new data about GNE expression patterns in human tissues. Our results suggest that there may be a link between tissue-specific pathology and isoform pattern in skeletal muscles, which could provide clues for the development of new treatment strategies for GNE myopathy.


Subject(s)
Distal Myopathies , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Humans , Muscle, Skeletal/metabolism , Mutation , N-Acetylneuraminic Acid/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
Neurol Sci ; 43(11): 6309-6321, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35904705

ABSTRACT

GNE myopathy is a hereditary muscle disorder characterized by muscle atrophy and weakness initially involving the lower distal extremities. The treatment of GNE myopathy mainly focuses on a sialic acid deficiency caused by a mutation in the GNE gene, but it has not achieved the expected effect. The main pathological features of GNE myopathy are myofiber atrophy and rimmed vacuoles, including accumulation of amyloid ß, which is mainly found in atrophic muscle fibers. Although the role of amyloid ß and other misfolded proteins on the nervous system has been widely recognized, the cause and process of the formation of amyloid ß in the pathological process of GNE myopathy are unclear. In addition, amyloid ß has been reported to be linked to quality control mechanisms of proteins, such as molecular chaperones, the ubiquitin-proteasome system, and the autophagy-lysosome system. Herein, we summarize the possible reasons for amyloid ß deposition and illustrate amyloid ß-mediated events in the cells and their role in muscle atrophy in GNE myopathy. This review represents an overview of amyloid ß and GNE myopathy that could help identify a potential mechanism and thereby a plausible therapeutic for the disease.


Subject(s)
Distal Myopathies , Muscular Diseases , Humans , Amyloid beta-Peptides/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , Muscular Atrophy , Muscle, Skeletal/pathology
5.
Neuropathology ; 41(1): 29-36, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33225515

ABSTRACT

UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) is necessary for sialic acid biosynthesis. GNE myopathy is caused by a defect in GNE, and hyposialylation is a key factor in the pathomechanism of GNE myopathy. Although candidates for evaluating hyposialylation have been reported, it is difficult to measure them in routine clinical practice. Sialylation is necessary for synthesis of various glycoproteins, including Krebs von den Lungen-6 (KL-6)/mucin 1 (MUC1). Here we report that KL-6/MUC1 is decreased in GNE myopathy. We observed that KL-6 levels were decreased in the serum of patients with GNE myopathy, and that KL-6 and MUC1-C were also decreased in muscle biopsy specimens from these patients. An immunofluorescent study revealed that KL-6 and MUC1-C were not present in the sarcolemma but were, instead, localized in rimmed vacuoles in specimens from patients with GNE myopathy. KL-6 is already used to detect lung diseases in clinical practice, and this glycoprotein may be a novel candidate for evaluating hyposialylation in GNE myopathy.


Subject(s)
Distal Myopathies/genetics , Distal Myopathies/metabolism , Mucin-1/metabolism , Multienzyme Complexes/genetics , Muscle, Skeletal/metabolism , Adult , Aged , Biomarkers/blood , Biomarkers/metabolism , Distal Myopathies/blood , Female , Humans , Male , Middle Aged , Mucin-1/blood , Young Adult
6.
Proc Natl Acad Sci U S A ; 115(28): E6566-E6575, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29946036

ABSTRACT

Myosin is a molecular motor indispensable for body movement and heart contractility. Apart from pure cardiomyopathy, mutations in MYH7 encoding slow/ß-cardiac myosin heavy chain also cause skeletal muscle disease with or without cardiac involvement. Mutations within the α-helical rod domain of MYH7 are mainly associated with Laing distal myopathy. To investigate the mechanisms underlying the pathology of the recurrent causative MYH7 mutation (K1729del), we have developed a Drosophila melanogaster model of Laing distal myopathy by genomic engineering of the Drosophila Mhc locus. Homozygous MhcK1728del animals die during larval/pupal stages, and both homozygous and heterozygous larvae display reduced muscle function. Flies expressing only MhcK1728del in indirect flight and jump muscles, and heterozygous MhcK1728del animals, were flightless, with reduced movement and decreased lifespan. Sarcomeres of MhcK1728del mutant indirect flight muscles and larval body wall muscles were disrupted with clearly disorganized muscle filaments. Homozygous MhcK1728del larvae also demonstrated structural and functional impairments in heart muscle, which were not observed in heterozygous animals, indicating a dose-dependent effect of the mutated allele. The impaired jump and flight ability and the myopathy of indirect flight and leg muscles associated with MhcK1728del were fully suppressed by expression of Abba/Thin, an E3-ligase that is essential for maintaining sarcomere integrity. This model of Laing distal myopathy in Drosophila recapitulates certain morphological phenotypic features seen in Laing distal myopathy patients with the recurrent K1729del mutation. Our observations that Abba/Thin modulates these phenotypes suggest that manipulation of Abba/Thin activity levels may be beneficial in Laing distal myopathy.


Subject(s)
Distal Myopathies , Drosophila Proteins/metabolism , Genetic Loci , Mutation , Myocardium/metabolism , Myosin Heavy Chains , Tripartite Motif Proteins , Animals , Disease Models, Animal , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Drosophila Proteins/genetics , Drosophila melanogaster , Homozygote , Humans , Myocardium/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics
7.
J Pathol ; 249(2): 182-192, 2019 10.
Article in English | MEDLINE | ID: mdl-31056746

ABSTRACT

Mutations in the Matrin 3 (MATR3) gene have been identified as a cause of amyotrophic lateral sclerosis (ALS) or vocal cord and pharyngeal weakness with distal myopathy (VCPDM). This study investigated the mechanism by which mutant MATR3 causes multisystem proteinopathy (MSP) including ALS and VCPDM. We first analyzed the muscle pathology of C57BL/6 mice injected with adeno-associated viruses expressing human WT or mutant (S85C) MATR3. We next generated transgenic mice that overexpress mutant (S85C) MATR3, driven by the CMV early enhancer/chicken ß-actin promoter, and evaluated their clinicopathological features. Intramuscular injection of viruses expressing WT and mutant MATR3 induced similar myogenic changes, including smaller myofibers with internal nuclei, and upregulated p62 and LC3-II. Mutant MATR3 transgenic mice showed decreased body weight and lower motor activity. Muscle histology demonstrated myopathic changes including fiber-size variation, internal nuclei and rimmed vacuoles. Spinal cord histology showed a reduced number of motor neurons, and activation of microglia and astrocytes. Comprehensive proteomic analyses of muscle demonstrated upregulation of proteins related to chaperones, stress response, protein degradation, and nuclear function. Overexpression of WT and mutant MATR3 similarly caused myotoxicity, recapitulating the clinicopathological features of MSP. These models will be helpful for analyzing MSP pathogenesis and for understanding the function of MATR3. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Distal Myopathies/genetics , Laryngeal Diseases/genetics , Muscle, Skeletal/metabolism , Mutation , Nuclear Matrix-Associated Proteins/genetics , Pharyngeal Diseases/genetics , RNA-Binding Proteins/genetics , Spinal Cord/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Dependovirus/genetics , Disease Models, Animal , Distal Myopathies/metabolism , Distal Myopathies/pathology , Distal Myopathies/physiopathology , Gait Analysis , Gene Transfer Techniques , Genetic Predisposition to Disease , Humans , Laryngeal Diseases/metabolism , Laryngeal Diseases/pathology , Laryngeal Diseases/physiopathology , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Motor Activity , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Nuclear Matrix-Associated Proteins/metabolism , Pharyngeal Diseases/metabolism , Pharyngeal Diseases/pathology , Pharyngeal Diseases/physiopathology , RNA-Binding Proteins/metabolism , Rotarod Performance Test , Sequestosome-1 Protein/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , Weight Loss
8.
Hum Mol Genet ; 26(16): 3081-3093, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28505249

ABSTRACT

Sialic acids are monosaccharides found in terminal sugar chains of cell surfaces and proteins; they have various biological functions and have been implicated in health and disease. Genetic defects of the GNE gene which encodes a critical bifunctional enzyme for sialic acid biosynthesis, lead to GNE myopathy, a disease manifesting with progressive muscle atrophy and weakness. The likely mechanism of disease is a lack of sialic acids. There remains, however, an unexplained link between hyposialylation and the muscle atrophy and weakness. In this study, we found that muscle proteins were highly modified by S-nitrosylation, and that oxidative stress-responsive genes were significantly upregulated, in hyposialylated muscles from human GNE myopathy patients and model mice. In both in vitro and in vivo models, the production of reactive oxygen species (ROS) was elevated with cellular hyposialylation, and increasing overall sialylation by extrinsic sialic acid intake reduced ROS and protein S-nitrosylation. More importantly, the antioxidant, oral N-acetylcysteine led to amelioration of the muscle atrophy and weakness in Gne mutant mice. Our data provide evidence of additional important function of sialic acids as a ROS scavenger in skeletal muscles, expanding our understanding on how sialic acid deficiency contributes to disease pathology, and identify oxidative stress as a therapeutic target in GNE myopathy.


Subject(s)
Distal Myopathies/metabolism , Distal Myopathies/pathology , N-Acetylneuraminic Acid/deficiency , Oxidative Stress/physiology , Acetylcysteine/metabolism , Acetylcysteine/therapeutic use , Animals , Disease Models, Animal , Humans , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , N-Acetylneuraminic Acid/metabolism , Reactive Oxygen Species/metabolism
9.
Muscle Nerve ; 60(1): 98-103, 2019 07.
Article in English | MEDLINE | ID: mdl-30990900

ABSTRACT

INTRODUCTION: UDP N-acetylglucosamine2-epimerase/N-acetylmannosamine-kinase (GNE) gene mutations can cause mostly autosomal-recessive myopathy with juvenile-onset known as hereditary inclusion-body myopathy (HIBM). METHODS: We describe a family of a patient showing an unusual HIBM with both vacuolar myopathy and myositis without quadriceps-sparing, hindering diagnosis. We show how genetic testing with functional assays, clinical transcriptome sequencing (RNA-seq) in particular, helped facilitate both the diagnosis and a better understanding of the genotype-phenotype relationship. RESULTS: We identified a novel 7.08 kb pathogenic deletion upstream of GNE using array comparative genomic hybridization (aCGH) and a common Val727Met variant. Using RNA-seq, we found only monoallelic (Val727Met-allele) expression, leading to ~50% GNE reduction in muscle. Importantly, α-dystroglycan is hypoglycosylated in the patient muscle, suggesting HIBM could be a "dystroglycanopathy." CONCLUSIONS: Our study shows the importance of considering aCGH for GNE-myopathies, and the potential of RNA-seq for faster, definitive molecular diagnosis of unusual myopathies. Muscle Nerve, 2019.


Subject(s)
Distal Myopathies/genetics , Multienzyme Complexes/genetics , Promoter Regions, Genetic/genetics , Comparative Genomic Hybridization , Distal Myopathies/diagnosis , Distal Myopathies/metabolism , Distal Myopathies/pathology , Dystroglycans/metabolism , Family , Gene Deletion , Glycosylation , Humans , Male , Molecular Diagnostic Techniques , Quadriceps Muscle/pathology , Sequence Analysis, RNA , Young Adult
10.
J Biol Chem ; 292(45): 18392-18407, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28939772

ABSTRACT

p97 is an essential ATPase associated with various cellular activities (AAA+) that functions as a segregase in diverse cellular processes, including the maintenance of proteostasis. p97 interacts with different cofactors that target it to distinct pathways; an important example is the deubiquitinase ataxin3, which collaborates with p97 in endoplasmic reticulum-associated degradation. However, the molecular details of this interaction have been unclear. Here, we characterized the binding of ataxin3 to p97, showing that ataxin3 binds with low-micromolar affinity to both wild-type p97 and mutants linked to degenerative disorders known as multisystem proteinopathy 1 (MSP1); we further showed that the stoichiometry of binding is one ataxin3 molecule per p97 hexamer. We mapped the binding determinants on each protein, demonstrating that ataxin3's p97/VCP-binding motif interacts with the inter-lobe cleft in the N-domain of p97. We also probed the nucleotide dependence of this interaction, confirming that ataxin3 and p97 associate in the presence of ATP and in the absence of nucleotide, but not in the presence of ADP. Our experiments suggest that an ADP-driven downward movement of the p97 N-terminal domain dislodges ataxin3 by inducing a steric clash between the D1-domain and ataxin3's C terminus. In contrast, MSP1 mutants of p97 bind ataxin3 irrespective of their nucleotide state, indicating a failure by these mutants to translate ADP binding into a movement of the N-terminal domain. Our model provides a mechanistic explanation for how nucleotides regulate the p97-ataxin3 interaction and why atypical cofactor binding is observed with MSP1 mutants.


Subject(s)
Ataxin-3/metabolism , Coenzymes/metabolism , Distal Myopathies/metabolism , Models, Molecular , Proteostasis Deficiencies/metabolism , Repressor Proteins/metabolism , Valosin Containing Protein/metabolism , Adenosine Diphosphate/chemistry , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Substitution , Ataxin-3/chemistry , Ataxin-3/genetics , Binding Sites , Binding, Competitive , Coenzymes/chemistry , Coenzymes/genetics , Crystallography, X-Ray , Databases, Protein , Distal Myopathies/enzymology , Distal Myopathies/genetics , Humans , Microscopy, Electron, Transmission , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Protein Multimerization , Proteostasis Deficiencies/enzymology , Proteostasis Deficiencies/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Repressor Proteins/chemistry , Repressor Proteins/genetics , Valosin Containing Protein/chemistry , Valosin Containing Protein/genetics
11.
Acta Neuropathol ; 135(1): 131-148, 2018 01.
Article in English | MEDLINE | ID: mdl-28780615

ABSTRACT

Mutations in the small heat shock protein B8 gene (HSPB8/HSP22) have been associated with distal hereditary motor neuropathy, Charcot-Marie-Tooth disease, and recently distal myopathy. It is so far not clear how mutant HSPB8 induces the neuronal and muscular phenotypes and if a common pathogenesis lies behind these diseases. Growing evidence points towards a role of HSPB8 in chaperone-associated autophagy, which has been shown to be a determinant for the clearance of poly-glutamine aggregates in neurodegenerative diseases but also for the maintenance of skeletal muscle myofibrils. To test this hypothesis and better dissect the pathomechanism of mutant HSPB8, we generated a new transgenic mouse model leading to the expression of the mutant protein (knock-in lines) or the loss-of-function (functional knock-out lines) of the endogenous protein Hspb8. While the homozygous knock-in mice developed motor deficits associated with degeneration of peripheral nerves and severe muscle atrophy corroborating patient data, homozygous knock-out mice had locomotor performances equivalent to those of wild-type animals. The distal skeletal muscles of the post-symptomatic homozygous knock-in displayed Z-disk disorganisation, granulofilamentous material accumulation along with Hspb8, αB-crystallin (HSPB5/CRYAB), and desmin aggregates. The presence of the aggregates correlated with reduced markers of effective autophagy. The sciatic nerve of the homozygous knock-in mice was characterized by low autophagy potential in pre-symptomatic and Hspb8 aggregates in post-symptomatic animals. On the other hand, the sciatic nerve of the homozygous knock-out mice presented a normal morphology and their distal muscle displayed accumulation of abnormal mitochondria but intact myofiber and Z-line organisation. Our data, therefore, suggest that toxic gain-of-function of mutant Hspb8 aggregates is a major contributor to the peripheral neuropathy and the myopathy. In addition, mutant Hspb8 induces impairments in autophagy that may aggravate the phenotype.


Subject(s)
Distal Myopathies/metabolism , Gain of Function Mutation , HSP20 Heat-Shock Proteins/genetics , HSP20 Heat-Shock Proteins/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myopathies, Structural, Congenital/metabolism , Peripheral Nervous System Diseases/metabolism , Animals , Atrophy/metabolism , Atrophy/pathology , Autophagy/physiology , Disease Models, Animal , Distal Myopathies/pathology , Female , Heat-Shock Proteins , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Molecular Chaperones , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/pathology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
12.
Hum Mol Genet ; 24(13): 3718-31, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25877298

ABSTRACT

Mutations in the extreme C-terminus of titin (TTN), situated in the sarcomeric M-band, cause tibial muscular dystrophy (TMD) and limb-girdle muscular dystrophy 2J (LGMD2J). The mutations ultimately cause a loss of C-terminal titin, including a binding site for the protease calpain 3 (CAPN3), and lead to a secondary CAPN3 deficiency in LGMD2J muscle. CAPN3 has been previously shown to bind C-terminal titin and to use it as a substrate in vitro. Interestingly, mutations in CAPN3 underlie limb-girdle muscular dystrophy 2A (LGMD2A). Here, we aimed to clarify the relationship of CAPN3 and M-band titin in normal and pathological muscle. In vitro analyses identified several CAPN3 cleavage sites in C-terminal titin that were defined by protein sequencing. Furthermore, cleavage products were detected in normal muscle extracts by western blotting and in situ by immunofluorescence microscopy. The TMD/LGMD2J mutation FINmaj proved to alter this processing in vitro, while binding of CAPN3 to mutant titin was preserved. Unexpectedly, the pathological loss of M-band titin due to TMD/LGMD2J mutations was found to be independent of CAPN3, whereas the involvement of ubiquitous calpains is likely. We conclude that proteolytic processing of C-terminal titin by CAPN3 may have an important role in normal muscle, and that this process is disrupted in LGMD2A and in TMD/LGMD2J due to CAPN3 deficiency and to the loss of C-terminal titin, respectively.


Subject(s)
Calpain/metabolism , Connectin/chemistry , Connectin/metabolism , Distal Myopathies/metabolism , Muscle Proteins/metabolism , Muscular Dystrophies, Limb-Girdle/enzymology , Protein Kinases/chemistry , Protein Kinases/metabolism , Amino Acid Motifs , Animals , Calpain/genetics , Connectin/genetics , Distal Myopathies/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/genetics , Muscle, Skeletal/enzymology , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/genetics , Protein Binding , Protein Kinases/genetics , Protein Processing, Post-Translational , Proteolysis
13.
Rev Neurol (Paris) ; 172(10): 587-593, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27638134

ABSTRACT

Distal myopathies and myofibrillar myopathies are both rare subcategories of muscle diseases. Myofibrillar myopathies are genetically heterogeneous group of diseases characterized by distinctive histopathology of abnormal protein aggregations and myofibrillar disintegration. All genes causing myofibrillar myopathy encode proteins that either reside in or associate with the Z-disc. Distal myopathies are also genetically heterogeneous muscular dystrophies in which muscle weakness presents distally in the feet and/or hands. A subgroup of distal myopathies, desminopathy, distal myotilinopathy, ZASPopathy and alpha-B crystallin-mutated distal myopathy, belong to myofibrillar myopathies and show similar pathological changes in muscle biopsies. Common features of these diseases are dominant inheritance and adult-onset of symptoms starting in the feet and slowly progressing to encompass other muscle groups. Cardiomyopathy is not a common feature in distal MFM myopathies.


Subject(s)
Distal Myopathies/pathology , Myofibrils/pathology , Distal Myopathies/genetics , Distal Myopathies/metabolism , Humans , Myofibrils/metabolism
14.
Am J Pathol ; 184(6): 1668-76, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24685690

ABSTRACT

Dysferlin is a membrane associated protein involved in vesicle trafficking and fusion. Defects in dysferlin result in limb-girdle muscular dystrophy type 2B and Miyoshi myopathy in humans and myopathy in A/J(dys-/-) and BLAJ mice, but the pathomechanism of the myopathy is not understood. Oil Red O staining showed many lipid droplets within the psoas and quadriceps muscles of dysferlin-deficient A/J(dys-/-) mice aged 8 and 12 months, and lipid droplets were also conspicuous within human myofibers from patients with dysferlinopathy (but not other myopathies). Electron microscopy of 8-month-old A/J(dys-/-) psoas muscles confirmed lipid droplets within myofibers and showed disturbed architecture of myofibers. In addition, the presence of many adipocytes was confirmed, and a possible role for dysferlin in adipocytes is suggested. Increased expression of mRNA for a gene involved in early lipogenesis, CCAAT/enhancer binding protein-δ, in 3-month-old A/J(dys-/-) quadriceps (before marked histopathology is evident), indicates early induction of lipogenesis/adipogenesis within dysferlin-deficient muscles. Similar results were seen for dysferlin-deficient BLAJ mice. These novel observations of conspicuous intermyofibrillar lipid and progressive adipocyte replacement in dysferlin-deficient muscles present a new focus for investigating the mechanisms that result in the progressive decline of muscle function in dysferlinopathies.


Subject(s)
Distal Myopathies/metabolism , Lipid Metabolism , Membrane Proteins/deficiency , Muscle Proteins/deficiency , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adolescent , Adult , Animals , Distal Myopathies/genetics , Distal Myopathies/pathology , Dysferlin , Female , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Middle Aged , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology
15.
J Recept Signal Transduct Res ; 35(4): 249-57, 2015.
Article in English | MEDLINE | ID: mdl-25510413

ABSTRACT

GNE Myopathy (GNEM) is a neuromuscular disorder caused by mutations in the GNE gene. It is a slowly progressive distal and proximal muscle weakness sparing the quadriceps. In this study, we applied our model of mutated M743T GNE enzyme skeletal muscle-cultured myoblasts and paired healthy controls to depict the pattern of signaling proteins controlling survival and/or apoptosis of the PI3K/AKT, BCL2, ARTS/XIAP pathways, examined the effects of metabolic changes/stimuli on their expression and activation, and their potential role in GNEM. Immunoblot analysis of the GNEM myoblasts indicated a notable increased level of activated PTEN and PDK1 and a trend of relative differences in the expression and activation of the examined signaling molecules with variability among the cultures. ANOVA analysis showed a highly significant interaction between the level of PTEN and the patients groups. In parallel, the interaction between the level of BCL2, BAX and PTEN with the specific PI3K/AKT inhibitor-LY294002 was highly significant for BCL2 and nearly significant for PTEN and BAX. The pattern of the ARTS/XIAP signaling proteins of GNEM and the paired controls was variable, with no significant differences between the two cell types. The response of the GNEM cells to the metabolic changes/stimuli: serum depletion and insulin challenge, as indicated by expression of selected signaling proteins, was variable and similar to the control cells. Taken together, our observations provide a clearer insight into specific signaling molecules influencing growth and survival of GNEM muscle cells.


Subject(s)
Distal Myopathies/metabolism , Distal Myopathies/pathology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myoblasts, Skeletal/metabolism , Myoblasts, Skeletal/pathology , Signal Transduction/physiology , Adult , Apoptosis , Case-Control Studies , Cell Survival , Cells, Cultured , Distal Myopathies/genetics , Female , Humans , Male , Middle Aged , Mutation , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Septins/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Young Adult
16.
Am J Hum Genet ; 88(6): 729-740, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21620354

ABSTRACT

Linkage analysis of the dominant distal myopathy we previously identified in a large Australian family demonstrated one significant linkage region located on chromosome 7 and encompassing 18.6 Mbp and 151 genes. The strongest candidate gene was FLNC because filamin C, the encoded protein, is muscle-specific and associated with myofibrillar myopathy. Sequencing of FLNC cDNA identified a c.752T>C (p.Met251Thr) mutation in the N-terminal actin-binding domain (ABD); this mutation segregated with the disease and was absent in 200 controls. We identified an Italian family with the same phenotype and found a c.577G>A (p.Ala193Thr) filamin C ABD mutation that segregated with the disease. Filamin C ABD mutations have not been described, although filamin A and filamin B ABD mutations cause multiple musculoskeletal disorders. The distal myopathy phenotype and muscle pathology in the two families differ from myofibrillar myopathies caused by filamin C rod and dimerization domain mutations because of the distinct involvement of hand muscles and lack of pathological protein aggregation. Thus, like the position of FLNA and B mutations, the position of the FLNC mutation determines disease phenotype. The two filamin C ABD mutations increase actin-binding affinity in a manner similar to filamin A and filamin B ABD mutations. Cell-culture expression of the c.752T>C (p.Met251)Thr mutant filamin C ABD demonstrated reduced nuclear localization as did mutant filamin A and filamin B ABDs. Expression of both filamin C ABD mutants as full-length proteins induced increased aggregation of filamin. We conclude filamin C ABD mutations cause a recognizable distal myopathy, most likely through increased actin affinity, similar to the pathological mechanism of filamin A and filamin B ABD mutations.


Subject(s)
Contractile Proteins/genetics , Distal Myopathies/genetics , Microfilament Proteins/genetics , Actins/metabolism , Adult , Aged , Australia , Chromosomes, Human, Pair 7/genetics , Contractile Proteins/metabolism , Distal Myopathies/metabolism , Distal Myopathies/pathology , Female , Filamins , Humans , Italy , Male , Microfilament Proteins/metabolism , Middle Aged , Mutation , Pedigree , Protein Structure, Tertiary/genetics
17.
FASEB J ; 27(8): 3217-28, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23657818

ABSTRACT

Myosin binding protein C (MyBP-C) is expressed in striated muscles, where it plays key roles in the modulation of actomyosin cross-bridges. Slow MyBP-C (sMyBP-C) consists of multiple variants sharing common domains but also containing unique segments within the NH2 and COOH termini. Two missense mutations in the NH2 terminus (W236R) and COOH terminus (Y856H) of sMyBP-C have been causally linked to the development of distal arthrogryposis-1 (DA-1), a severe skeletal muscle disorder. Using a combination of in vitro binding and motility assays, we show that the COOH terminus mediates binding of sMyBP-C to thick filaments, while the NH2 terminus modulates the formation of actomyosin cross-bridges in a variant-specific manner. Consistent with this, a recombinant NH2-terminal peptide that excludes residues 34-59 reduces the sliding velocity of actin filaments past myosin heads from 9.0 ± 1.3 to 5.7 ± 1.0 µm/s at 0.1 µM, while a recombinant peptide that excludes residues 21-59 fails to do so. Notably, the actomyosin regulatory properties of sMyBP-C are completely abolished by the presence of the DA-1 mutations. In summary, our studies are the first to show that the NH2 and COOH termini of sMyBP-C have distinct functions, which are regulated by differential splicing, and are compromized by the presence of missense point mutations linked to muscle disease.


Subject(s)
Actomyosin/metabolism , Arthrogryposis/metabolism , Carrier Proteins/metabolism , Distal Myopathies/metabolism , Actins/chemistry , Actins/metabolism , Actomyosin/chemistry , Alternative Splicing , Amino Acid Substitution , Animals , Arthrogryposis/genetics , Binding Sites/genetics , Blotting, Far-Western , Carrier Proteins/chemistry , Carrier Proteins/genetics , Distal Myopathies/genetics , Humans , Mice , Models, Biological , Models, Molecular , Muscle, Skeletal/metabolism , Mutation , Myosins/chemistry , Myosins/metabolism , Protein Binding , Protein Structure, Tertiary
18.
J Neuromuscul Dis ; 11(5): 905-917, 2024.
Article in English | MEDLINE | ID: mdl-38875046

ABSTRACT

Background: GNE Myopathy is a unique recessive neuromuscular disorder characterized by adult-onset, slowly progressive distal and proximal muscle weakness, caused by mutations in the GNE gene which is a key enzyme in the biosynthesis of sialic acid. To date, the precise pathophysiology of the disease is not well understood and no reliable animal model is available. Gne KO is embryonically lethal in mice. Objective: To gain insights into GNE function in muscle, we have generated an inducible muscle Gne KO mouse. To minimize the contribution of the liver to the availability of sialic acid to muscle via the serum, we have also induced combined Gne KO in liver and muscle. Methods: A mouse carrying loxp sequences flanking Gne exon3 was generated by Crispr/Cas9 and bred with a human skeletal actin (HSA) promoter driven CreERT mouse. Gne muscle knock out was induced by tamoxifen injection of the resulting homozygote GneloxpEx3loxp/HSA Cre mouse. Liver Gne KO was induced by systemic injection of AAV8 vectors carrying the Cre gene driven by the hepatic specific promoter of the thyroxine binding globulin gene. Results: Characterization of these mice for a 12 months period showed no significant changes in their general behaviour, motor performance, muscle mass and structure in spite of a dramatic reduction in sialic acid content in both muscle and liver. Conclusions: We conclude that post weaning lack of Gne and sialic acid in muscle and liver have no pathologic effect in adult mice. These findings could reflect a strong interspecies versatility, but also raise questions about the loss of function hypothesis in Gne Myopathy. If these findings apply to humans they have a major impact on therapeutic strategies.


Subject(s)
Disease Models, Animal , Liver , Mice, Knockout , Muscle, Skeletal , Animals , Mice , Muscle, Skeletal/metabolism , Liver/metabolism , Distal Myopathies/genetics , Distal Myopathies/metabolism , Carbohydrate Epimerases/genetics , Carbohydrate Epimerases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , N-Acetylneuraminic Acid/metabolism
19.
Stem Cell Res ; 80: 103491, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39047410

ABSTRACT

Variants in MYH7 cause cardiomyopathies as well as myosin storage myopathy and Laing early-onset distal myopathy (MPD1). MPD1 is characterized by muscle weakness and atrophy usually beginning in the lower legs. Here, we generated iPSC lines from lymphoblastoid cells of three unrelated individuals heterozygous for the most common MPD1-causing variant; p.Lys1617del. iPSC lines showed typical morphology, expressed pluripotency markers, demonstrated trilineage differentiation potential, and had a normal karyotype. These lines represent the first iPSCs derived from MPD1 patients and complement existing MPD1 animal models. They can provide in vitro platforms to better understand and model MPD1 pathomechanisms and test therapies.


Subject(s)
Cardiac Myosins , Distal Myopathies , Induced Pluripotent Stem Cells , Myosin Heavy Chains , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Distal Myopathies/genetics , Distal Myopathies/pathology , Distal Myopathies/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Male , Female , Cell Line , Cell Differentiation , Adult
20.
J Clin Invest ; 134(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38690726

ABSTRACT

Proline substitutions within the coiled-coil rod region of the ß-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.


Subject(s)
Amino Acid Substitution , Distal Myopathies , Proline , Animals , Mice , Humans , Proline/genetics , Proline/metabolism , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Mutation, Missense , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/chemistry , Female , Male , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology
SELECTION OF CITATIONS
SEARCH DETAIL