Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Eur J Neurol ; 28(2): 729-731, 2021 02.
Article in English | MEDLINE | ID: mdl-33047465

ABSTRACT

α-methylacyl-CoA racemase (AMACR) deficiency is a rare disorder, affecting peroxisomal metabolism of pristanic acid, with ten published adult cases. We describe an AMACR deficiency case with a clinical presentation dominated by episodic hyperCKaemia, suggesting that myopathic features of AMACR should be considered.


Subject(s)
Lipid Metabolism, Inborn Errors , Racemases and Epimerases , Adult , Coenzyme A , Humans , Racemases and Epimerases/genetics
2.
Acta Neuropathol ; 139(3): 565-582, 2020 03.
Article in English | MEDLINE | ID: mdl-31897643

ABSTRACT

Protein O-glucosyltransferase 1 (POGLUT1) activity is critical for the Notch signaling pathway, being one of the main enzymes responsible for the glycosylation of the extracellular domain of Notch receptors. A biallelic mutation in the POGLUT1 gene has been reported in one family as the cause of an adult-onset limb-girdle muscular dystrophy (LGMD R21; OMIM# 617232). As the result of a collaborative international effort, we have identified the first cohort of 15 patients with LGMD R21, from nine unrelated families coming from different countries, providing a reliable phenotype-genotype and mechanistic insight. Patients carrying novel mutations in POGLUT1 all displayed a clinical picture of limb-girdle muscle weakness. However, the age at onset was broadened from adult to congenital and infantile onset. Moreover, we now report that the unique muscle imaging pattern of "inside-to-outside" fatty degeneration observed in the original cases is indeed a defining feature of POGLUT1 muscular dystrophy. Experiments on muscle biopsies from patients revealed a remarkable and consistent decrease in the level of the NOTCH1 intracellular domain, reduction of the pool of satellite cells (SC), and evidence of α-dystroglycan hypoglycosylation. In vitro biochemical and cell-based assays suggested a pathogenic role of the novel POGLUT1 mutations, leading to reduced enzymatic activity and/or protein stability. The association between the POGLUT1 variants and the muscular phenotype was established by in vivo experiments analyzing the indirect flight muscle development in transgenic Drosophila, showing that the human POGLUT1 mutations reduced its myogenic activity. In line with the well-known role of the Notch pathway in the homeostasis of SC and muscle regeneration, SC-derived myoblasts from patients' muscle samples showed decreased proliferation and facilitated differentiation. Together, these observations suggest that alterations in SC biology caused by reduced Notch1 signaling result in muscular dystrophy in LGMD R21 patients, likely with additional contribution from α-dystroglycan hypoglycosylation. This study settles the muscular clinical phenotype linked to POGLUT1 mutations and establishes the pathogenic mechanism underlying this muscle disorder. The description of a specific imaging pattern of fatty degeneration and muscle pathology with a decrease of α-dystroglycan glycosylation provides excellent tools which will help diagnose and follow up LGMD R21 patients.


Subject(s)
Dystroglycans/metabolism , Glucosyltransferases/genetics , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Animals , Animals, Genetically Modified , Drosophila melanogaster , Female , Genetic Association Studies , Glycosylation , Humans , Male , Muscle, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Mutation , Pedigree , Satellite Cells, Skeletal Muscle/pathology
3.
Eur J Neurol ; 27(11): 2257-2266, 2020 11.
Article in English | MEDLINE | ID: mdl-32558070

ABSTRACT

BACKGROUND AND PURPOSE: Very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD) is a hereditary disorder of mitochondrial long-chain fatty acid oxidation that has variable presentations, including exercise intolerance, cardiomyopathy and liver disease. The aim of this study was to describe the clinical and genetic manifestations of six patients with adult-onset VLCADD. METHODS: In this study, the clinical, pathological and genetic findings of six adult patients (four from Iran and two from Serbia) with VLCADD and their response to treatment are described. RESULTS: The median (range) age of patients at first visit was 31 (27-38) years, and the median (range) age of onset was 26.5 (19-33) years. Parental consanguinity was present for four patients. Four patients had a history of rhabdomyolysis, and the recorded CK level ranged between 67 and 90 000 IU/l. Three patients had a history of exertional myalgia, and one patient had a non-fluctuating weakness. Through next-generation sequencing analysis, we identified six cases with variants in the ACADVL gene and a confirmed diagnosis of VLCADD. Of the total six variants identified, five were missense, and one was a novel frameshift mutation identified in two unrelated individuals. Two variants were novel, and three were previously reported. We treated the patients with a combination of L-carnitine, Coenzyme Q10 and riboflavin. Three patients responded favorably to the treatment. CONCLUSION: Adult-onset VLCADD is a rare entity with various presentations. Patients may respond favorably to a cocktail of L-carnitine, Coenzyme Q10, and riboflavin.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Lipid Metabolism, Inborn Errors , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Adult , Congenital Bone Marrow Failure Syndromes , Female , Humans , Male , Mitochondrial Diseases , Muscular Diseases , Young Adult
4.
Eur J Neurol ; 27(12): 2604-2615, 2020 12.
Article in English | MEDLINE | ID: mdl-32697863

ABSTRACT

BACKGROUND AND PURPOSE: The aim was to define the radiological picture of facioscapulohumeral muscular dystrophy 2 (FSHD2) in comparison with FSHD1 and to explore correlations between imaging and clinical/molecular data. METHODS: Upper girdle and/or lower limb muscle magnetic resonance imaging scans of 34 molecularly confirmed FSHD2 patients from nine European neuromuscular centres were analysed. T1-weighted and short-tau inversion recovery (STIR) sequences were used to evaluate the global pattern and to assess the extent of fatty replacement and muscle oedema. RESULTS: The most frequently affected muscles were obliquus and transversus abdominis, semimembranosus, soleus and gluteus minimus in the lower limbs; trapezius, serratus anterior, latissimus dorsi and pectoralis major in the upper girdle. Iliopsoas, popliteus, obturator internus and tibialis posterior in the lower limbs and subscapularis, spinati, sternocleidomastoid and levator scapulae in the upper girdle were the most spared. Asymmetry and STIR hyperintensities were consistent features. The pattern of muscle involvement was similar to that of FSHD1, and the combined involvement of trapezius, abdominal and hamstring muscles, together with complete sparing of iliopsoas and subscapularis, was detected in 91% of patients. Peculiar differences were identified in a rostro-caudal gradient, a predominant involvement of lower limb muscles compared to the upper girdle, and in the higher percentage of STIR hyperintensities in FSHD2. CONCLUSION: This multicentre study defines the pattern of muscle involvement in FSHD2, providing useful information for diagnostics and clinical trial design. Both similarities and differences between FSHD1 and FSHD2 were detected, which is also relevant to better understand the pathogenic mechanisms underlying the FSHD-related disease spectrum.


Subject(s)
Muscular Dystrophy, Facioscapulohumeral , Humans , Lower Extremity , Magnetic Resonance Imaging , Muscle, Skeletal/diagnostic imaging , Muscular Dystrophy, Facioscapulohumeral/diagnostic imaging , Muscular Dystrophy, Facioscapulohumeral/genetics
5.
Acta Neuropathol ; 138(6): 1013-1031, 2019 12.
Article in English | MEDLINE | ID: mdl-31463572

ABSTRACT

MSTO1 encodes a cytosolic mitochondrial fusion protein, misato homolog 1 or MSTO1. While the full genotype-phenotype spectrum remains to be explored, pathogenic variants in MSTO1 have recently been reported in a small number of patients presenting with a phenotype of cerebellar ataxia, congenital muscle involvement with histologic findings ranging from myopathic to dystrophic and pigmentary retinopathy. The proposed underlying pathogenic mechanism of MSTO1-related disease is suggestive of impaired mitochondrial fusion secondary to a loss of function of MSTO1. Disorders of mitochondrial fusion and fission have been shown to also lead to mitochondrial DNA (mtDNA) depletion, linking them to the mtDNA depletion syndromes, a clinically and genetically diverse class of mitochondrial diseases characterized by a reduction of cellular mtDNA content. However, the consequences of pathogenic variants in MSTO1 on mtDNA maintenance remain poorly understood. We present extensive phenotypic and genetic data from 12 independent families, including 15 new patients harbouring a broad array of bi-allelic MSTO1 pathogenic variants, and we provide functional characterization from seven MSTO1-related disease patient fibroblasts. Bi-allelic loss-of-function variants in MSTO1 manifest clinically with a remarkably consistent phenotype of childhood-onset muscular dystrophy, corticospinal tract dysfunction and early-onset non-progressive cerebellar atrophy. MSTO1 protein was not detectable in the cultured fibroblasts of all seven patients evaluated, suggesting that pathogenic variants result in a loss of protein expression and/or affect protein stability. Consistent with impaired mitochondrial fusion, mitochondrial networks in fibroblasts were found to be fragmented. Furthermore, all fibroblasts were found to have depletion of mtDNA ranging from 30 to 70% along with alterations to mtDNA nucleoids. Our data corroborate the role of MSTO1 as a mitochondrial fusion protein and highlight a previously unrecognized link to mtDNA regulation. As impaired mitochondrial fusion is a recognized cause of mtDNA depletion syndromes, this novel link to mtDNA depletion in patient fibroblasts suggests that MSTO1-deficiency should also be considered a mtDNA depletion syndrome. Thus, we provide mechanistic insight into the disease pathogenesis associated with MSTO1 mutations and further define the clinical spectrum and the natural history of MSTO1-related disease.


Subject(s)
Cell Cycle Proteins/genetics , Cerebellar Diseases/genetics , Cytoskeletal Proteins/genetics , DNA, Mitochondrial , Mitochondrial Diseases/genetics , Muscular Dystrophies/genetics , Mutation , Adolescent , Adult , Atrophy , Cells, Cultured , Cerebellar Diseases/diagnostic imaging , Cerebellar Diseases/pathology , Cerebellar Diseases/physiopathology , Child , DNA Copy Number Variations , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Male , Middle Aged , Mitochondrial Diseases/diagnostic imaging , Mitochondrial Diseases/pathology , Mitochondrial Diseases/physiopathology , Muscles/pathology , Muscular Dystrophies/diagnostic imaging , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Phenotype , Young Adult
6.
BMC Pediatr ; 19(1): 131, 2019 04 25.
Article in English | MEDLINE | ID: mdl-31023296

ABSTRACT

BACKGROUND: Adolescents with DMD treated with chronic high dose GC therapy typically have profound pubertal delay. Testosterone, the main circulating androgen in men, promotes virilisation and growth with associated accrual of fat-free muscle mass and bone mineral content. Testosterone therapy is routinely used to mimic the normal stages of pubertal development in patients with hypogonadotrophic hypogonadism, androgen deficiency secondary to testicular disease and in constitutional delay of growth and puberty (CDGP). Improved life expectancy in DMD has meant that more adolescents are eligible for testosterone supplementation but there is little objective data regarding the impact of this treatment on muscle structure and function, bone integrity and overall well-being. METHODS: This is a single centre observational clinical trial (NCT02571205) that aims to follow the progress of 15 adolescents with Duchenne muscular dystrophy and delayed puberty as they are managed with incremental testosterone therapy to induce puberty. Subjects will all be treated with a steadily increasing dose of testosterone administered by injection every 4 weeks and data will be collected to help us determine the effectiveness and tolerability of the described treatment regimen. We will use the data to explore the effects of testosterone on pubertal development, growth, muscle strength and function, bone mineral density, body composition with a detailed record of any adverse events. We will also carry out interviews to explore the boys' views on the tolerability of the regimen. The study will last for 27 months in total for each participant. DISCUSSION: Our experience has indicated that testosterone treatment in adolescents with DMD is liked and well tolerated but we have not collected objective data on a specific treatment regimen and there is no current consensus. Testosterone supplementation is not part of the standard of care of pubertal delay in DMD but inclusion in future protocols may be appropriate depending on the results of this trial. TRIAL REGISTRATION: EudraCT Number: 2015-003195-68. Research Registry & References: Clinical trials.gov- NCT02571205 (registered 8/10/15).


Subject(s)
Glucocorticoids/adverse effects , Muscle Strength/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Puberty, Delayed/drug therapy , Testosterone/administration & dosage , Adolescent , Body Composition/drug effects , Bone Density/drug effects , Child , Dose-Response Relationship, Drug , Drug Administration Schedule , Follow-Up Studies , Glucocorticoids/administration & dosage , Humans , Injections, Intramuscular , Male , Muscular Dystrophy, Duchenne/complications , Muscular Dystrophy, Duchenne/diagnosis , Patient Satisfaction , Prospective Studies , Treatment Outcome , United Kingdom
7.
Clin Genet ; 90(2): 166-70, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27409453

ABSTRACT

Four and a half LIM protein 1 (FHL1/SLIM1) has recently been identified as the causative gene mutated in four distinct diseases affecting skeletal muscle that have overlapping features, including reducing body myopathy, X-linked myopathy, X-linked dominant scapuloperoneal myopathy and Emery-Dreifuss muscular dystrophy. FHL1 localises to the sarcomere and the sarcolemma and is believed to participate in muscle growth and differentiation as well as in sarcomere assembly. We describe in this case report a boy with a deletion of the entire FHL1 gene who is now 15 years of age and presented with muscle hypertrophy, reduced subcutaneous fat, rigid spine and short stature. This case is the first, to our knowledge, with a complete loss of the FHL1 protein and MAP7D3 in combination. It supports the theory that dominant negative effects (accumulation of cytotoxic-mutated FHL1 protein) worsen the pathogenesis. It extends the phenotype of FHL1-related myopathies and should prompt future testing in undiagnosed patients who present with unexplained muscle hypertrophy, contractures and rigid spine, particularly if male.


Subject(s)
Gene Deletion , Hypertrophy/genetics , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Microtubule-Associated Proteins/genetics , Muscle Proteins/genetics , Muscular Diseases/genetics , Spine/pathology , Subcutaneous Fat/pathology , Adolescent , Gene Expression , Humans , Hypertrophy/pathology , Intracellular Signaling Peptides and Proteins/deficiency , LIM Domain Proteins/deficiency , Male , Microtubule-Associated Proteins/deficiency , Muscle Proteins/deficiency , Muscular Diseases/pathology , Phenotype , Spine/metabolism , Subcutaneous Fat/metabolism
8.
Neuromuscul Disord ; 33(3): 241-249, 2023 03.
Article in English | MEDLINE | ID: mdl-36753800

ABSTRACT

Pregnancy and birth in women with neuromuscular conditions has been associated with more rapid disease progression and obstetric complications. This study assessed the impact of functional status and specific diagnosis on patient reported pregnancy and birth outcomes in 26 genetic neuromuscular diseases. Pregnancy and birth outcomes were collected through electronic patient questionnaires and analysed by mobility group and diagnosis. Free text responses were grouped into themes. 721 pregnancies were reported by 305 women. Miscarriage (21% of pregnancies), caesarean delivery (38% of births) and instrumental vaginal delivery (19% of births) were all more frequent in respondents than in the general population (p<0.05), and were more common in those who were non-ambulant at conception than other mobility levels (p <0.05). Falls occurred during 42% of pregnancies and a deterioration in muscle strength during 43%. There was not an increased incidence of maternal complications, apart from maternal hypertension which was more common in limb girdle muscular dystrophy 2A/R1 (35%) and myotonic dystrophy (24%). Patients offered specific practical advice to prospective mothers. Women with neuromuscular conditions have a more complex antenatal and perinatal course than unaffected women. Prenatal counselling, specialist obstetric review and additional occupational therapy support should be considered.


Subject(s)
Neuromuscular Diseases , Pregnancy Outcome , Pregnancy , Humans , Female , Prospective Studies , Delivery, Obstetric , Patient Reported Outcome Measures
9.
Ann Neurol ; 68(5): 717-26, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20839240

ABSTRACT

OBJECTIVE: Centronuclear myopathy (CNM) is a rare congenital myopathy characterized by prominence of central nuclei on muscle biopsy. CNM has been associated with mutations in MTM1, DNM2, and BIN1 but many cases remain genetically unresolved. RYR1 encodes the principal sarcoplasmic reticulum calcium release channel and has been implicated in various congenital myopathies. We investigated whether RYR1 mutations cause CNM. METHODS: We sequenced the entire RYR1 coding sequence in 24 patients with a diagnosis of CNM from South Africa (n = 14) and Europe (n = 10) and identified mutations in 17 patients. The most common genotypes featured compound heterozygosity for RYR1 missense mutations and mutations resulting in reduced protein expression, including intronic splice site and frameshift mutations. RESULTS: The high incidence in South African patients (n = 12/14) in conjunction with recurrent RYR1 mutations associated with common haplotypes suggested the presence of founder effects. In addition to central nuclei, prominent histopathological findings included (often multiple) internalized nuclei and type 1 fiber predominance and hypotrophy with relative type 2 hypertrophy. Although cores were not typically seen on oxidative stains, electron microscopy revealed subtle abnormalities in most cases. External ophthalmoplegia, proximal weakness, and bulbar involvement were prominent clinical findings. INTERPRETATION: Our findings expand the range of RYR1-related phenotypes and suggest RYR1 mutations as a common cause of congenital myopathies with central nuclei. Corresponding to recent observations in X-linked CNM, these findings indicate disturbed assembly and/or malfunction of the excitation-contraction machinery as a key mechanism in CNM and related myopathies.


Subject(s)
Muscle, Skeletal/pathology , Myopathies, Structural, Congenital/etiology , Ryanodine Receptor Calcium Release Channel/genetics , Adolescent , Child , Child, Preschool , Europe , Genotype , Heterozygote , Humans , Male , Mutation , Myopathies, Structural, Congenital/diagnosis , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Phenotype , Ryanodine Receptor Calcium Release Channel/metabolism , South Africa
11.
Neuromuscul Disord ; 31(12): 1259-1265, 2021 12.
Article in English | MEDLINE | ID: mdl-34702655

ABSTRACT

Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy in childhood. It is associated with progressive muscle function decline and premature death. Long-term oral glucocorticoid use slows muscle weakness but is associated with several side effects including delayed puberty. This study assessed the impact of a 2-year incremental intramuscular testosterone regimen on quality of life (QoL) in a cohort of 15 adolescents with DMD. The Pediatric Quality of Life Inventory (PedsQL) Neuromuscular module was used to assess QoL and was completed by parent-child dyads. Semi-structured interviews were carried out to understand patient views on testosterone therapy. QoL scores increased in 10 of the 15 participants during treatment, with a mean total PedsQL score of 74.6 pre-treatment v 80.2 post treatment (p = 0.04). This was supported by comments in the semi-structured interviews. Parent-reported PedsQL scores were lower than their child's post treatment (p = 0.007). Testosterone therapy for pubertal induction was associated with an improvement in QoL and the observed physical changes during puberty played an important role. Low self-esteem was also a prevailing theme. This data supports the inclusion of testosterone therapy for pubertal induction as a Standard of Care.


Subject(s)
Androgens/pharmacology , Dwarfism/drug therapy , Muscular Dystrophy, Duchenne/drug therapy , Patient Reported Outcome Measures , Puberty/drug effects , Quality of Life , Testosterone/pharmacology , Adolescent , Androgens/administration & dosage , Child , Dwarfism/psychology , Humans , Male , Muscular Dystrophy, Duchenne/psychology , Parents , Self Concept , Testosterone/administration & dosage
12.
Brain ; 132(Pt 1): 147-55, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19015158

ABSTRACT

Mutations in COL6A1, COL6A2 and COL6A3, the genes which encode the extra-cellular matrix component collagen VI, lead to Bethlem myopathy and Ullrich congenital muscular dystrophy (UCMD). Although the Col6a1(-/-) null mouse has an extremely mild neuromuscular phenotype, a mitochondrial defect has been demonstrated, linked to dysregulation of the mitochondrial permeability transition pore (PTP) opening. This finding has been replicated in UCMD muscle cells in culture, providing justification for a clinical trial using cyclosporine A, an inhibitor of PTP opening. We investigated whether PTP dysregulation could be detected in UCMD fibroblasts (the predominant source of muscle collagen VI), in myoblast cells from patients with other diseases and its response to rescue agents other than collagen VI. Although we confirm the presence of PTP dysregulation in muscle-derived cultures from two UCMD patients, fibroblasts from the same patients and the majority of fibroblasts from other well-characterized UCMD patients behave normally. PTP dysregulation is found in limb girdle muscular dystrophy (LGMD) type 2B myoblasts but not in myoblasts from patients with Bethlem myopathy, merosin-deficient congenital muscular dystrophy, LGMD2A, Duchenne muscular dystrophy and Leigh syndrome. In addition to rescue by cyclosporine A and collagen VI, this cellular phenotype was also rescued by other extra-cellular matrix constituents (laminin and collagen I). As the muscle derived cultures demonstrating PTP dysregulation shared poor growth in culture and lack of desmin labelling, we believe that PTP dysregulation may be a particular characteristic of the state of these cells in culture and is not specific to the collagen VI defect, and can in any case be rescued by a range of extra-cellular matrix components. Further work is needed on the relationship of PTP dysregulation with UCMD pathology.


Subject(s)
Cyclosporine/pharmacology , Mitochondria/physiology , Muscular Dystrophies/pathology , Adolescent , Cells, Cultured , Child , Child, Preschool , Collagen Type VI/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Infant , Infant, Newborn , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/physiology , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/metabolism , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Rhodamines , Skin/metabolism , Young Adult
13.
BMC Genomics ; 10: 451, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19775440

ABSTRACT

BACKGROUND: The freshwater snail Lymnaea stagnalis (L. stagnalis) has served as a successful model for studies in the field of Neuroscience. However, a serious drawback in the molecular analysis of the nervous system of L. stagnalis has been the lack of large-scale genomic or neuronal transcriptome information, thereby limiting the use of this unique model. RESULTS: In this study, we report 7,712 distinct EST sequences (median length: 847 nucleotides) of a normalized L. stagnalis central nervous system (CNS) cDNA library, resulting in the largest collection of L. stagnalis neuronal transcriptome data currently available. Approximately 42% of the cDNAs can be translated into more than 100 consecutive amino acids, indicating the high quality of the library. The annotated sequences contribute 12% of the predicted transcriptome size of 20,000. Surprisingly, approximately 37% of the L. stagnalis sequences only have a tBLASTx hit in the EST library of another snail species Aplysia californica (A. californica) even using a low stringency e-value cutoff at 0.01. Using the same cutoff, approximately 67% of the cDNAs have a BLAST hit in the NCBI non-redundant protein and nucleotide sequence databases (nr and nt), suggesting that one third of the sequences may be unique to L. stagnalis. Finally, using the same cutoff (0.01), more than half of the cDNA sequences (54%) do not have a hit in nematode, fruitfly or human genome data, suggesting that the L. stagnalis transcriptome is significantly different from these species as well. The cDNA sequences are enriched in the following gene ontology functional categories: protein binding, hydrolase, transferase, and catalytic enzymes. CONCLUSION: This study provides novel molecular insights into the transcriptome of an important molluscan model organism. Our findings will contribute to functional analyses in neurobiology, and comparative evolutionary biology. The L. stagnalis CNS EST database is available at http://www.Lymnaea.org/.


Subject(s)
Central Nervous System/metabolism , Expressed Sequence Tags , Gene Expression Profiling , Lymnaea/genetics , Amino Acid Sequence , Animals , Aplysia/genetics , Biomphalaria/genetics , Chromosome Mapping , Comparative Genomic Hybridization , Computational Biology , Gene Library , Molecular Sequence Data , Phylogeny , Sequence Alignment
14.
Dev Cell ; 1(5): 717-24, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11709191

ABSTRACT

Muscle-eye-brain disease (MEB) is an autosomal recessive disorder characterized by congenital muscular dystrophy, ocular abnormalities, and lissencephaly. Mammalian O-mannosyl glycosylation is a rare type of protein modification that is observed in a limited number of glycoproteins of brain, nerve, and skeletal muscle. Here we isolated a human cDNA for protein O-mannose beta-1,2-N-acetylglucosaminyltransferase (POMGnT1), which participates in O-mannosyl glycan synthesis. We also identified six independent mutations of the POMGnT1 gene in six patients with MEB. Expression of most frequent mutation revealed a great loss of the enzymatic activity. These findings suggest that interference in O-mannosyl glycosylation is a new pathomechanism for muscular dystrophy as well as neuronal migration disorder.


Subject(s)
Cell Movement , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Muscular Dystrophies/enzymology , Muscular Dystrophies/genetics , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Point Mutation/genetics , Amino Acid Sequence , Base Sequence , Blotting, Western , Cell Line , Child, Preschool , Cloning, Molecular , DNA Mutational Analysis , Female , Gene Expression , Glycosyltransferases/chemistry , Humans , Male , Molecular Sequence Data , Muscular Dystrophies/pathology , Mutagenesis, Site-Directed , N-Acetylglucosaminyltransferases/chemistry , Pedigree , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Substrate Specificity
15.
J Cell Biol ; 119(5): 1183-91, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1447296

ABSTRACT

Dystrophin, the protein product of the Duchenne muscular dystrophy (DMD) gene locus, is expressed on the muscle fiber surface. One key to further understanding of the cellular function of dystrophin would be extended knowledge about its subcellular organization. We have shown that dystrophin molecules are not uniformly distributed over the humen, rat, and mouse skeletal muscle fiber surface using three independent methods. Incubation of single-teased muscle fibers with antibodies to dystrophin revealed a network of denser transversal rings (costameres) and finer longitudinal interconnections. Double staining of longitudinal semithin cryosections for dystrophin and alpha-actinin showed spatial juxtaposition of the costameres to the Z bands. Where peripheral myonuclei precluded direct contact of dystrophin to the Z bands the organization of dystrophin was altered into lacunae harboring the myonucleus. These lacunae were surrounded by a dystrophin ring and covered by a more uniform dystrophin veil. Mechanical skinning of single-teased fibers revealed tighter mechanical connection of dystrophin to the plasma membrane than to the underlying internal domain of the muscle fiber. The entire dystrophin network remained preserved in its structure on isolated muscle sarcolemma and identical in appearance to the pattern observed on teased fibers. Therefore, connection of defined areas of plasma membrane or its constituents such as ion channels to single sarcomeres might be a potential function exerted by dystrophin alone or in conjunction with other submembrane cytoskeletal proteins.


Subject(s)
Cell Compartmentation , Cell Membrane/chemistry , Dystrophin/isolation & purification , Muscles/chemistry , Muscles/ultrastructure , Muscular Dystrophies/pathology , Actinin/immunology , Actinin/isolation & purification , Animals , Cell Membrane/ultrastructure , Dystrophin/immunology , Dystrophin/ultrastructure , Fluorescent Antibody Technique , Humans , Mice , Mice, Inbred C57BL , Muscular Dystrophy, Animal/pathology , Rats , Rats, Inbred Strains , Rats, Wistar
16.
J Cell Biol ; 139(2): 375-85, 1997 Oct 20.
Article in English | MEDLINE | ID: mdl-9334342

ABSTRACT

Genetic defects in a number of components of the dystrophin-glycoprotein complex (DGC) lead to distinct forms of muscular dystrophy. However, little is known about how alterations in the DGC are manifested in the pathophysiology present in dystrophic muscle tissue. One hypothesis is that the DGC protects the sarcolemma from contraction-induced damage. Using tracer molecules, we compared sarcolemmal integrity in animal models for muscular dystrophy and in muscular dystrophy patient samples. Evans blue, a low molecular weight diazo dye, does not cross into skeletal muscle fibers in normal mice. In contrast, mdx mice, a dystrophin-deficient animal model for Duchenne muscular dystrophy, showed significant Evans blue accumulation in skeletal muscle fibers. We also studied Evans blue dispersion in transgenic mice bearing different dystrophin mutations, and we demonstrated that cytoskeletal and sarcolemmal attachment of dystrophin might be a necessary requirement to prevent serious fiber damage. The extent of dye incorporation in transgenic mice correlated with the phenotypic severity of similar dystrophin mutations in humans. We furthermore assessed Evans blue incorporation in skeletal muscle of the dystrophia muscularis (dy/dy) mouse and its milder allelic variant, the dy2J/dy2J mouse, animal models for congenital muscular dystrophy. Surprisingly, these mice, which have defects in the laminin alpha2-chain, an extracellular ligand of the DGC, showed little Evans blue accumulation in their skeletal muscles. Taken together, these results suggest that the pathogenic mechanisms in congenital muscular dystrophy are different from those in Duchenne muscular dystrophy, although the primary defects originate in two components associated with the same protein complex.


Subject(s)
Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Muscular Dystrophy, Animal/pathology , Sarcolemma/pathology , Animals , Dystrophin/biosynthesis , Dystrophin/deficiency , Dystrophin/genetics , Evans Blue , Humans , Mice , Mice, Inbred mdx , Mice, Mutant Strains , Mice, Transgenic , Mutagenesis , Mutation , Myocardium/pathology , Serum Albumin/analysis
17.
J Cell Biol ; 145(1): 153-65, 1999 Apr 05.
Article in English | MEDLINE | ID: mdl-10189375

ABSTRACT

The dystrophin-glycoprotein complex (DGC) is a multisubunit complex that spans the muscle plasma membrane and forms a link between the F-actin cytoskeleton and the extracellular matrix. The proteins of the DGC are structurally organized into distinct subcomplexes, and genetic mutations in many individual components are manifested as muscular dystrophy. We recently identified a unique tetraspan-like dystrophin-associated protein, which we have named sarcospan (SPN) for its multiple sarcolemma spanning domains (Crosbie, R.H., J. Heighway, D.P. Venzke, J.C. Lee, and K.P. Campbell. 1997. J. Biol. Chem. 272:31221-31224). To probe molecular associations of SPN within the DGC, we investigated SPN expression in normal muscle as a baseline for comparison to SPN's expression in animal models of muscular dystrophy. We show that, in addition to its sarcolemma localization, SPN is enriched at the myotendinous junction (MTJ) and neuromuscular junction (NMJ), where it is a component of both the dystrophin- and utrophin-glycoprotein complexes. We demonstrate that SPN is preferentially associated with the sarcoglycan (SG) subcomplex, and this interaction is critical for stable localization of SPN to the sarcolemma, NMJ, and MTJ. Our experiments indicate that assembly of the SG subcomplex is a prerequisite for targeting SPN to the sarcolemma. In addition, the SG- SPN subcomplex functions to stabilize alpha-dystroglycan to the muscle plasma membrane. Taken together, our data provide important information about assembly and function of the SG-SPN subcomplex.


Subject(s)
Carrier Proteins/metabolism , Cytoskeletal Proteins/physiology , Dystrophin/physiology , Membrane Glycoproteins/physiology , Membrane Proteins/metabolism , Muscular Dystrophy, Animal/metabolism , Neoplasm Proteins , Amino Acid Sequence , Animals , Biological Transport , CHO Cells , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Cricetinae , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Dystroglycans , Dystrophin/deficiency , Dystrophin/genetics , Humans , Hydrogen-Ion Concentration , Macromolecular Substances , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Knockout , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Neuromuscular Junction/metabolism , Rabbits , Sarcoglycans , Sarcolemma/metabolism , Sarcolemma/ultrastructure , Sequence Alignment , Species Specificity , Tendons/metabolism , Transfection , Utrophin
18.
J Cell Biol ; 142(6): 1461-71, 1998 Sep 21.
Article in English | MEDLINE | ID: mdl-9744877

ABSTRACT

Limb-girdle muscular dystrophy type 2D (LGMD 2D) is an autosomal recessive disorder caused by mutations in the alpha-sarcoglycan gene. To determine how alpha-sarcoglycan deficiency leads to muscle fiber degeneration, we generated and analyzed alpha-sarcoglycan- deficient mice. Sgca-null mice developed progressive muscular dystrophy and, in contrast to other animal models for muscular dystrophy, showed ongoing muscle necrosis with age, a hallmark of the human disease. Sgca-null mice also revealed loss of sarcolemmal integrity, elevated serum levels of muscle enzymes, increased muscle masses, and changes in the generation of absolute force. Molecular analysis of Sgca-null mice demonstrated that the absence of alpha-sarcoglycan resulted in the complete loss of the sarcoglycan complex, sarcospan, and a disruption of alpha-dystroglycan association with membranes. In contrast, no change in the expression of epsilon-sarcoglycan (alpha-sarcoglycan homologue) was observed. Recombinant alpha-sarcoglycan adenovirus injection into Sgca-deficient muscles restored the sarcoglycan complex and sarcospan to the membrane. We propose that the sarcoglycan-sarcospan complex is requisite for stable association of alpha-dystroglycan with the sarcolemma. The Sgca-deficient mice will be a valuable model for elucidating the pathogenesis of sarcoglycan deficient limb-girdle muscular dystrophies and for the development of therapeutic strategies for this disease.


Subject(s)
Cytoskeletal Proteins/deficiency , Membrane Glycoproteins/deficiency , Muscular Dystrophy, Animal/etiology , Neoplasm Proteins , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/biosynthesis , Carrier Proteins/physiology , Cytoskeletal Proteins/genetics , DNA, Complementary , Disease Progression , Dystrophin/metabolism , Gene Transfer Techniques , Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Muscle Contraction , Muscular Dystrophy, Animal/physiopathology , Sarcoglycans , Sarcolemma/metabolism
19.
Cardiovasc Res ; 79(4): 652-61, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18495669

ABSTRACT

AIMS: As oral corticosteroids have a beneficial effect on muscle strength in Duchenne muscular dystrophy, it has been suggested that they may also be a useful treatment in the pathologically related sarcoglycanopathies. The delta-sarcoglycan-deficient mouse (Sgcd-null) is a model for both limb girdle muscular dystrophy 2F (LGMD2F) and dilated cardiomyopathy. METHODS AND RESULTS: To study the effect of oral corticosteroids on cardiac function, we treated 8-week-old Sgcd-null mice with prednisolone (1.5 mg/kg body weight/day orally) for 8 weeks. In vivo cardiac function was assessed by pressure-volume loops using a conductance catheter. We found a well-compensated cardiomyopathy at baseline in Sgcd-null mice with decreased myocardial contractility, increased preload, and decreased afterload, maintaining a high cardiac output. Cardiac haemodynamics, surprisingly, did not improve in prednisolone-treated mice, but instead deteriorated with evidence of ventricular stiffening. On histology, after steroid treatment there was increased myocardial cell damage and increased myocardial fibrosis. CONCLUSION: Prednisolone led to a decompensation of cardiac haemodynamics in Sgcd-null mice and induced additional cardiac damage. On the basis of these findings, although mouse models may not completely replicate the human situation for LGMD2F, we conclude that careful cardiac monitoring is clearly indicated in patients on long-term corticosteroids.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Cardiomyopathy, Dilated/drug therapy , Hemodynamics/drug effects , Myocardium/pathology , Prednisolone/pharmacology , Sarcoglycans/deficiency , Ventricular Function, Left/drug effects , Administration, Oral , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/adverse effects , Adrenergic beta-Agonists/administration & dosage , Animals , Body Weight/drug effects , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Dobutamine/administration & dosage , Fibrosis , Infusions, Intravenous , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardium/metabolism , Prednisolone/administration & dosage , Prednisolone/adverse effects , RNA, Messenger/metabolism , Sarcoglycans/genetics , Stroke Volume/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ventricular Pressure/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL