Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
Add more filters

Publication year range
1.
Gene Ther ; 24(9): 514-519, 2017 09.
Article in English | MEDLINE | ID: mdl-28561813

ABSTRACT

Spinal muscular atrophy is an autosomal recessive neurodegenerative disorder characterized by progressive muscle wasting and loss of muscle function due to severe motor neuron dysfunction, secondary to mutations in the survival motor neuron 1 (SMN1) gene. A second neighboring centromeric gene, SMN2, is intact in all patients but contains a C-to-T variation in exon 7 that affects a splice enhancer and determines exclusion of exon 7 in the majority of its transcript, leading to an unstable protein that cannot substitute for mutant SMN1. Following successful studies on disease models and intensive studies on SMN functions in the past decade, SMN upregulation targeting SMN2, has been suggested as a possible therapeutic approach. Recently, we have witnessed an historical turning point with the first disease-modifying treatment receiving Food and Drug Administration approval and now being available to patients also outside the clinical trial. This innovative treatment is an antisense oligonucleotide, which, administered intrathecally, is able to increase exon 7 inclusion in the majority of the SMN2 mRNA and increase the production of fully functional SMN protein. Alternative advanced therapies, such as viral vector mediated gene therapy and orally available small molecules, are also showing promising results in early clinical trial phases.


Subject(s)
Genetic Therapy/methods , Muscular Atrophy, Spinal/therapy , Animals , Clinical Trials as Topic , Genetic Therapy/adverse effects , Humans , Muscular Atrophy, Spinal/genetics , Oligonucleotides, Antisense/administration & dosage , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
2.
J Neurol Neurosurg Psychiatry ; 86(8): 873-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25430934

ABSTRACT

BACKGROUND: The international Inherited Neuropathy Consortium (INC) was created with the goal of obtaining much needed natural history data for patients with Charcot-Marie-Tooth (CMT) disease. We analysed clinical and genetic data from patients in the INC to determine the distribution of CMT subtypes and the clinical impairment associated with them. METHODS: We analysed data from 1652 patients evaluated at 13 INC centres. The distribution of CMT subtypes and pathogenic genetic mutations were determined. The disease burden of all the mutations was assessed by the CMT Neuropathy Score (CMTNS) and CMT Examination Score (CMTES). RESULTS: 997 of the 1652 patients (60.4%) received a genetic diagnosis. The most common CMT subtypes were CMT1A/PMP22 duplication, CMT1X/GJB1 mutation, CMT2A/MFN2 mutation, CMT1B/MPZ mutation, and hereditary neuropathy with liability to pressure palsy/PMP22 deletion. These five subtypes of CMT accounted for 89.2% of all genetically confirmed mutations. Mean CMTNS for some but not all subtypes were similar to those previously reported. CONCLUSIONS: Our findings confirm that large numbers of patients with a representative variety of CMT subtypes have been enrolled and that the frequency of achieving a molecular diagnosis and distribution of the CMT subtypes reflects those previously reported. Measures of severity are similar, though not identical, to results from smaller series. This study confirms that it is possible to assess patients in a uniform way between international centres, which is critical for the planned natural history study and future clinical trials. These data will provide a representative baseline for longitudinal studies of CMT. CLINICAL TRIAL REGISTRATION: ID number NCT01193075.


Subject(s)
Charcot-Marie-Tooth Disease/classification , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Charcot-Marie-Tooth Disease/genetics , Charcot-Marie-Tooth Disease/pathology , Charcot-Marie-Tooth Disease/physiopathology , Connexins/genetics , Cost of Illness , Cross-Sectional Studies , Female , GTP Phosphohydrolases/genetics , Humans , Male , Mitochondrial Proteins/genetics , Mutation/genetics , Myelin P0 Protein/genetics , Myelin Proteins/genetics , Nuclear Proteins , Proteins/genetics , Gap Junction beta-1 Protein
3.
Child Care Health Dev ; 39(3): 449-55, 2013 May.
Article in English | MEDLINE | ID: mdl-22676208

ABSTRACT

BACKGROUND: Gene-modifying trials offer hope for improvement in chronic paediatric disorders, but they may also lead to disappointment and have an adverse emotional effect on families. This study aimed to examine emotional impact on participants in a paediatric exon-skipping trial. METHODS: Nineteen male children with Duchenne muscular dystrophy (DMD), and their parents, taking part in a dose-ranging study of an i.v. administered morpholino splice-switching oligomer (which can restore the reading frame in DMD and induce dystrophin expression) underwent a psychosocial/psychiatric examination at trial entry. Emotional impact was assessed at trial completion using questionnaires. RESULTS: The mean child age was 8.9 years (SD 2.1); 13(68%) were attending mainstream school. Most families were well adjusted psychosocially at trial entry. Post-trial median child emotional impact scores were 5/10 (n= 18), but impact was rated as positive by 6/14 (42%), neutral/mixed by 5 (35%) and negative by 3 (21%). Median post-trial psychosocial/psychiatric change scores in children and parents were minimal. Actual post-trial negative impact was statistically significantly associated with higher expected impact at trial entry, at which time the families of the three children displaying actual negative impact reported higher family stress levels in combination with a variety of other psychosocial risks factors. CONCLUSIONS: In carefully selected families with low levels of psychosocial stress/distress at trial entry, and with good support from paediatric research units (including psychiatric input when required), genetic trials in progressive disorders such as DMD can have a predominantly positive or neutral emotional impact. Nevertheless, negative impact is reported by a minority of families and possible psychosocial predictors deserving further scrutiny have been identified.


Subject(s)
Affective Symptoms/etiology , Genetic Therapy/psychology , Muscular Dystrophy, Duchenne/therapy , Child , Dose-Response Relationship, Drug , Family Health , Genetic Therapy/methods , Humans , Male , Morpholinos/genetics , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/psychology , Oligonucleotides/administration & dosage , Oligonucleotides/therapeutic use , Parents/psychology , Psychiatric Status Rating Scales , Psychometrics , Stress, Psychological/etiology
4.
Nat Genet ; 29(1): 17-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528383

ABSTRACT

One form of congenital muscular dystrophy, rigid spine syndrome (MIM 602771), is a rare neuromuscular disorder characterized by early rigidity of the spine and respiratory insufficiency. A locus on 1p35-36 (RSMD1) was recently found to segregate with rigid spine muscular dystrophy 1 (ref. 1). Here we refine the locus and find evidence of linkage disequilibrium associated with SEPN1, which encodes the recently described selenoprotein N (ref. 2). Our identification and analysis of mutations in SEPN1 is the first description of a selenoprotein implicated in a human disease.


Subject(s)
Lung Diseases/genetics , Muscle Proteins/genetics , Muscular Dystrophies/genetics , Mutation , Spine/physiopathology , Amino Acid Sequence , Animals , Chromosome Mapping , Chromosomes, Human, Pair 1 , Humans , Molecular Sequence Data , Muscle Proteins/chemistry , Muscular Dystrophies/congenital , Polymerase Chain Reaction , Polymorphism, Single-Stranded Conformational , Selenoproteins , Sequence Homology, Amino Acid
5.
Nat Genet ; 21(3): 285-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10080180

ABSTRACT

Emery-Dreifuss muscular dystrophy (EDMD) is characterized by early contractures of elbows and Achilles tendons, slowly progressive muscle wasting and weakness, and a cardiomyopathy with conduction blocks which is life-threatening. Two modes of inheritance exist, X-linked (OMIM 310300) and autosomal dominant (EDMD-AD; OMIM 181350). EDMD-AD is clinically identical to the X-linked forms of the disease. Mutations in EMD, the gene encoding emerin, are responsible for the X-linked form. We have mapped the locus for EDMD-AD to an 8-cM interval on chromosome 1q11-q23 in a large French pedigree, and found that the EMD phenotype in four other small families was potentially linked to this locus. This region contains the lamin A/C gene (LMNA), a candidate gene encoding two proteins of the nuclear lamina, lamins A and C, produced by alternative splicing. We identified four mutations in LMNA that co-segregate with the disease phenotype in the five families: one nonsense mutation and three missense mutations. These results are the first identification of mutations in a component of the nuclear lamina as a cause of inherited muscle disorder. Together with mutations in EMD (refs 5,6), they underscore the potential importance of the nuclear envelope components in the pathogenesis of neuromuscular disorders.


Subject(s)
Muscular Dystrophies/genetics , Mutation , Nuclear Proteins/genetics , Amino Acid Sequence , Cloning, Molecular , Deoxyribonuclease HpaII/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Exons , Female , Genes, Dominant , Haplotypes , Humans , Immunohistochemistry , Lamin Type A , Lamins , Male , Microsatellite Repeats , Molecular Sequence Data , Muscular Dystrophy, Emery-Dreifuss , Myocardium/metabolism , Myocardium/pathology , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Pedigree , Sequence Analysis, DNA , Sequence Homology, Amino Acid
6.
Nat Genet ; 29(1): 75-7, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11528396

ABSTRACT

Classic spinal muscular atrophy (SMA) is caused by mutations in the telomeric copy of SMN1. Its product is involved in various cellular processes, including cytoplasmic assembly of spliceosomal small nuclear ribonucleoproteins, pre-mRNA processing and activation of transcription. Spinal muscular atrophy with respiratory distress (SMARD) is clinically and genetically distinct from SMA. Here we demonstrate that SMARD type 1 (SMARD1) results from mutations in the gene encoding immunoglobulin micro-binding protein 2 (IGHMBP2; on chromosome 11q13.2-q13.4). In six SMARD1 families, we detected three recessive missense mutations (exons 5, 11 and 12), two nonsense mutations (exons 2 and 5), one frameshift deletion (exon 5) and one splice donor-site mutation (intron 13). Mutations in mouse Ighmbp2 (ref. 14) have been shown to be responsible for spinal muscular atrophy in the neuromuscular degeneration (nmd) mouse, whose phenotype resembles the SMARD1 phenotype. Like the SMN1 product, IGHMBP2 colocalizes with the RNA-processing machinery in both the cytoplasm and the nucleus. Our results show that IGHMBP2 is the second gene found to be defective in spinal muscular atrophy, and indicate that IGHMBP2 and SMN share common functions important for motor neuron maintenance and integrity in mammals.


Subject(s)
Carrier Proteins/genetics , DNA-Binding Proteins , Muscular Atrophy, Spinal/genetics , Mutation, Missense , Respiratory Distress Syndrome, Newborn/genetics , Transcription Factors , Amino Acid Sequence , Animals , Base Sequence , Carrier Proteins/chemistry , Chromosomes, Human, Pair 11 , DNA Primers , Female , Humans , Infant, Newborn , Male , Mice , Molecular Sequence Data , Pedigree , Sequence Homology, Amino Acid
7.
Nat Genet ; 23(2): 208-12, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508519

ABSTRACT

Muscle contraction results from the force generated between the thin filament protein actin and the thick filament protein myosin, which causes the thick and thin muscle filaments to slide past each other. There are skeletal muscle, cardiac muscle, smooth muscle and non-muscle isoforms of both actin and myosin. Inherited diseases in humans have been associated with defects in cardiac actin (dilated cardiomyopathy and hypertrophic cardiomyopathy), cardiac myosin (hypertrophic cardiomyopathy) and non-muscle myosin (deafness). Here we report that mutations in the human skeletal muscle alpha-actin gene (ACTA1) are associated with two different muscle diseases, 'congenital myopathy with excess of thin myofilaments' (actin myopathy) and nemaline myopathy. Both diseases are characterized by structural abnormalities of the muscle fibres and variable degrees of muscle weakness. We have identified 15 different missense mutations resulting in 14 different amino acid changes. The missense mutations in ACTA1 are distributed throughout all six coding exons, and some involve known functional domains of actin. Approximately half of the patients died within their first year, but two female patients have survived into their thirties and have children. We identified dominant mutations in all but 1 of 14 families, with the missense mutations being single and heterozygous. The only family showing dominant inheritance comprised a 33-year-old affected mother and her two affected and two unaffected children. In another family, the clinically unaffected father is a somatic mosaic for the mutation seen in both of his affected children. We identified recessive mutations in one family in which the two affected siblings had heterozygous mutations in two different exons, one paternally and the other maternally inherited. We also identified de novo mutations in seven sporadic probands for which it was possible to analyse parental DNA.


Subject(s)
Actins/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Myopathies, Nemaline/genetics , Adolescent , Adult , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Child , Child, Preschool , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Family Health , Female , Humans , Infant , Male , Molecular Sequence Data , Mutation , Point Mutation , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational , Sequence Analysis, DNA , Sequence Homology, Amino Acid
8.
Neuromuscul Disord ; 33(6): 476-483, 2023 06.
Article in English | MEDLINE | ID: mdl-37207382

ABSTRACT

Eteplirsen is FDA-approved for the treatment of Duchenne muscular dystrophy (DMD) in exon 51 skip-amenable patients. Previous studies in boys > 4 years of age indicate eteplirsen is well tolerated and attenuates pulmonary and ambulatory decline compared with matched natural history cohorts. Here the safety, tolerability and pharmacokinetics of eteplirsen in boys aged 6-48 months is evaluated. In this open-label, multicenter, dose-escalation study (NCT03218995), boys with a confirmed mutation of the DMD gene amenable to exon 51 skipping (Cohort 1: aged 24-48 months, n = 9; Cohort 2: aged 6 to < 24 months, n = 6) received ascending doses (2, 4, 10, 20, 30 mg/kg) of once-weekly eteplirsen intravenously over 10 weeks, continuing at 30 mg/kg up to 96 weeks. Endpoints included safety (primary) and pharmacokinetics (secondary). All 15 participants completed the study. Eteplirsen was well tolerated with no treatment-related discontinuations, deaths or evidence of kidney toxicity. Most treatment-emergent adverse events were mild; most common were pyrexia, cough, nasopharyngitis, vomiting, and diarrhea. Eteplirsen pharmacokinetics were consistent between both cohorts and with previous clinical experience in boys with DMD > 4 years of age. These data support the safety and tolerability of eteplirsen at the approved 30-mg/kg dose in boys as young as 6 months old.


Subject(s)
Muscular Dystrophy, Duchenne , Male , Humans , Child, Preschool , Infant , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Morpholinos/therapeutic use , Exons , Mutation , Dystrophin/genetics
9.
J Med Genet ; 48(10): 660-668, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21931168

ABSTRACT

OBJECTIVES: Homoplasmic maternally inherited, m.14674T>C or m. 14674T>G mt-tRNA(Glu) mutations have recently been identified in reversible infantile cytochrome c oxidase deficiency (or 'benign COX deficiency'). This study sought other genetic defects that may give rise to similar presentations. PATIENTS: Eight patients from seven families with clinicopathological features of infantile reversible cytochrome c oxidase deficiency were investigated. METHODS: The study reviewed the diagnostic features and performed molecular genetic analyses of mitochondrial DNA and nuclear encoded candidate genes. RESULTS: Patients presented with subacute onset of profound hypotonia, feeding difficulties and lactic acidosis within the first months of life. Although recovery was remarkable, a mild myopathy persisted into adulthood. Histopathological findings in muscle included increased lipid and/or glycogen content, ragged-red and COX negative fibres. Biochemical studies suggested more generalised abnormalities than pure COX deficiency. Clinical improvement was reflected by normalisation of lactic acidosis and histopathological abnormalities. The m.14674T>C mt-tRNA(Glu) mutation was identified in four families, but none had the m. 14674T>G mutation. Furthermore, in two families pathogenic mutations were also found in the nuclear TRMU gene which has not previously been associated with this phenotype. In one family, the genetic aetiology still remains unknown. CONCLUSIONS: Benign COX deficiency is better described as 'reversible infantile respiratory chain deficiency'. It is genetically heterogeneous, and patients not carrying the m.14674T>C or T>G mt-tRNA(Glu) mutations may have mutations in the TRMU gene. Diagnosing this disorder at the molecular level is a significant advance for paediatric neurologists and intensive care paediatricians, enabling them to select children with an excellent prognosis for continuing respiratory support from those with severe mitochondrial presentation in infancy.


Subject(s)
Cytochrome-c Oxidase Deficiency/genetics , Acidosis, Lactic/genetics , Acidosis, Lactic/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , Brain/pathology , Child , Child, Preschool , Cytochrome-c Oxidase Deficiency/metabolism , Cytochrome-c Oxidase Deficiency/pathology , Electron Transport Complex IV/genetics , Face/pathology , Family , Female , Genetic Heterogeneity , Histocytochemistry , Humans , Infant , Infant, Newborn , Liver/pathology , Magnetic Resonance Imaging , Male , Mitochondrial Proteins/genetics , Molecular Sequence Data , Muscle Hypotonia , Muscle, Skeletal/pathology , Mutation/genetics , Sequence Alignment , tRNA Methyltransferases/genetics
10.
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(4): 300-309, 2021 04.
Article in English | MEDLINE | ID: mdl-33752934

ABSTRACT

Nusinersen (NUS), the first treatment approved for Spinal Muscular Atrophy type 1 (SMA1), was made available in the UK for SMA1 through the Expanded Access Program (EAP) in 2017. The Great Ormond Street Respiratory (GSR) score was developed as an objective respiratory assessment for children with SMA1 during their treatment. Aims: Track respiratory status of SMA1 children over the course of Nusinersen treatment and compare GSR scores amongst SMA1 sub-types. Single centre study on SMA1 patients using the GSR score at set time points: prior to first NUS dose; 2 weeks post end of loading doses; 2 weeks post-subsequent doses. GSR score ranges 1-28, being 1-9 = Stable minimal support, thorough to 23-28 = Poor reserve with maximum support. 20 SMA1 children underwent NUS treatment between January 2017 - November 2018. Median age of diagnosis was 5.0 months. NUS started at median of 9.57 months. From 5th dose onwards, GSR scores were significantly lower for Type 1C patients compared to Type 1B By month 18, irrespective of subtypes, the whole cohort appears to stabilise GSR Scores. As treatment duration increases, an overall stabilisation of respiratory status across the cohort was observed. Further longitudinal studies are needed to validate the GSR.


Subject(s)
Oligonucleotides/therapeutic use , Respiratory Function Tests/methods , Spinal Muscular Atrophies of Childhood/drug therapy , Cohort Studies , Female , Humans , Infant , Male , Respiratory System/drug effects
12.
AJNR Am J Neuroradiol ; 42(1): 167-172, 2021 01.
Article in English | MEDLINE | ID: mdl-33122211

ABSTRACT

BACKGROUND AND PURPOSE: Walker-Warburg syndrome, muscle-eye-brain disease, and Fukuyama congenital muscular dystrophy are α-dystroglycan-related muscular disorders associated with brain malformations and eye abnormalities in which no structural inner ear abnormality has been described radiologically. We collected patients from 6 tertiary pediatric hospitals and reported the radiologic features and frequency of inner ear dysplasias. MATERIALS AND METHODS: Patients previously diagnosed clinicoradiologically with Walker-Warburg syndrome, muscle-eye-brain disease, or Fukuyama congenital muscular dystrophy were included. We recorded the pathogenic variant, when available. Brain MR imaging and/or CT findings were reviewed in consensus, and inner ear anomalies were classified according to previous description in the literature. We then correlated the clinicoradiologic phenotype with the inner ear phenotype. RESULTS: Thirteen patients fulfilled the criteria for the Walker-Warburg syndrome phenotype, 8 for muscle-eye-brain disease, and 3 for Fukuyama congenital muscular dystrophy. A dysplastic cochlea was demonstrated in 17/24. The most frequent finding was a pronounced cochlear hypoplasia type 4 with a very small anteriorly offset turn beyond the normal-appearing basal turn (12/13 patients with Walker-Warburg syndrome and 1/11 with muscle-eye-brain disease or Fukuyama congenital muscular dystophy). Two of 8 patients with muscle-eye-brain disease, 1/3 with Fukuyama congenital muscular dystrophy, and 1/13 with Walker-Warburg syndrome showed a less severe cochlear hypoplasia type 4. The remaining patients without Walker-Warburg syndrome were healthy. The vestibule and lateral semicircular canals of all patients were normal. Cranial nerve VIII was present in all patients with diagnostic MR imaging. CONCLUSIONS: Most patients with the severe α-dystroglycanopathy Walker-Warburg syndrome phenotype have a highly characteristic cochlear hypoplasia type 4. Patients with the milder variants, muscle-eye-brain disease and Fukuyama congenital muscular dystrophy, more frequently have a normal cochlea or milder forms of hypoplasia.


Subject(s)
Cochlea/abnormalities , Walker-Warburg Syndrome/pathology , Adolescent , Child , Child, Preschool , Dystroglycans/genetics , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging/methods , Male , Neuroimaging , Phenotype , Walker-Warburg Syndrome/complications , Walker-Warburg Syndrome/genetics , Young Adult
13.
Neuropathol Appl Neurobiol ; 36(4): 265-74, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20002311

ABSTRACT

AIMS: The quantification of protein levels in muscle biopsies is of particular relevance in the diagnostic process of neuromuscular diseases, but is difficult to assess in cases of partial protein deficiency, particularly when information on protein localization is required. The combination of immunohistochemistry and Western blotting is often used in these cases, but is not always possible if the sample is scarce. We therefore sought to develop a method to quantify relative levels of sarcolemma-associated proteins using digitally captured images of immunolabelled sections of skeletal muscle. METHODS: To validate our relative quantification method, we labelled dystrophin and other sarcolemmal proteins in transverse sections of muscle biopsies taken from Duchenne muscular dystrophy and Becker muscular dystrophy patients, a manifesting carrier of Duchenne muscular dystrophy and normal controls. RESULTS: Using this method to quantify relative sarcolemmal protein abundance, we were able to accurately distinguish between the different patients on the basis of the relative amount of dystrophin present. CONCLUSIONS: This comparative method adds value to techniques that are already part of the diagnostic process and can be used with minimal variation of the standardized protocols, without using extra amounts of valuable biopsy samples. Comparative quantification of sarcolemmal proteins on immunostained muscle sections will be of use to establish both the abundance and localization of the protein. Moreover, it can be applied to assess the efficacy of experimental therapies where only partial restoration or upregulation of the protein may occur.


Subject(s)
Immunohistochemistry/methods , Membrane Proteins/metabolism , Muscle, Skeletal/metabolism , Case-Control Studies , Child , Diagnosis, Differential , Dystrophin/metabolism , Female , Heterozygote , Humans , Image Processing, Computer-Assisted , Male , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Sarcolemma/metabolism , Scoliosis/metabolism , Software
14.
Brain ; 132(Pt 2): 439-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19155270

ABSTRACT

Mutations in fukutin related protein (FKRP) are responsible for a common group of muscular dystrophies ranging from adult onset limb girdle muscular dystrophies to severe congenital forms with associated structural brain involvement, including Muscle Eye Brain disease. A common feature of these disorders is the variable reduction in the glycosylation of skeletal muscle alpha-dystroglycan. In order to gain insight into the pathogenesis and clinical variability, we have generated two lines of mice, the first containing a missense mutation and a neomycin cassette, FKRP-Neo(Tyr307Asn) and the second containing the FKRP(Tyr307Asn) mutation alone. We have previously associated this missense mutation with a severe muscle-eye-brain phenotype in several families. Homozygote Fkrp-Neo(Tyr307Asn) mice die soon after birth and show a reduction in the laminin-binding epitope of alpha-dystroglycan in muscle, eye and brain, and have reduced levels of FKRP transcript. Homozygous Fkrp(Tyr307Asn) mice showed no discernible phenotype up to 6 months of age, contrary to the severe clinical course observed in patients with the same mutation. These results suggest the generation of a mouse model for FKRP related muscular dystrophy requires a knock-down rather than a knock-in strategy in order to give rise to a disease phenotype.


Subject(s)
Muscular Dystrophies/genetics , Mutation, Missense , Proteins/genetics , Animals , Blotting, Southern , Cell Movement , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chimera , Dystroglycans/metabolism , Female , Gene Targeting , Genotype , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Models, Animal , Muscular Dystrophies/metabolism , Muscular Dystrophies/physiopathology , Pentosyltransferases , Phenotype , Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transferases
15.
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
16.
J Neurol Neurosurg Psychiatry ; 80(7): 706-14, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19531685

ABSTRACT

Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and limb girdle muscular dystrophies (LGMD) represent a significant proportion of paediatric and adult neuromuscular neurology practice. The proactive symptom-based multidisciplinary team (MDT) management and access to non-invasive ventilation have enabled improved survival into adulthood. Nevertheless the severe disability imposed by conditions such as DMD poses a challenge for successful transition of care and management for paediatric and adult neurology teams. DMD is discussed in detail as a paradigm illustrating diagnosis, management and role for different pharmacological interventions to improve survival, but also challenges in adulthood care, and cutting-edge therapies. LGMDs are much rarer than DMD and BMD, and in addition there is a significant genetic and clinical heterogeneity, which leads to diagnostic difficulties. The clinical and laboratory diagnostic features of seven LGMD subtypes are summarised, and their allelic "non-limb girdle" phenotypes are tabulated to illustrate the theme of one gene causing multiple clinical phenotypes, with the aim of refining the clinician's diagnostic approach. The lessons learnt from DMD MDT management to improve survival are broadly applicable to LGMDs with severe motor disability/multisystem complications.


Subject(s)
Adrenal Cortex Hormones/therapeutic use , Muscular Dystrophies/diagnosis , Muscular Dystrophies/therapy , Physical Therapy Modalities , Sleep Apnea Syndromes/therapy , Adult , Child , Humans , Muscular Dystrophies/complications , Muscular Dystrophies/genetics , Muscular Dystrophies/physiopathology , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/therapy , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/therapy , Sleep Apnea Syndromes/etiology , Treatment Outcome
17.
Postgrad Med J ; 85(1009): 622-30, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19892898

ABSTRACT

Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD) and limb girdle muscular dystrophies (LGMD) represent a significant proportion of paediatric and adult neuromuscular neurology practice. The proactive symptom-based multidisciplinary team (MDT) management and access to non-invasive ventilation have enabled improved survival into adulthood. Nevertheless the severe disability imposed by conditions such as DMD poses a challenge for successful transition of care and management for paediatric and adult neurology teams. DMD is discussed in detail as a paradigm illustrating diagnosis, management and role for different pharmacological interventions to improve survival, but also challenges in adulthood care, and cutting-edge therapies. LGMDs are much rarer than DMD and BMD, and in addition there is a significant genetic and clinical heterogeneity, which leads to diagnostic difficulties. The clinical and laboratory diagnostic features of seven LGMD subtypes are summarised, and their allelic "non-limb girdle" phenotypes are tabulated to illustrate the theme of one gene causing multiple clinical phenotypes, with the aim of refining the clinician's diagnostic approach. The lessons learnt from DMD MDT management to improve survival are broadly applicable to LGMDs with severe motor disability/multisystem complications.

18.
Neuromuscul Disord ; 29(4): 261-268, 2019 04.
Article in English | MEDLINE | ID: mdl-30852071

ABSTRACT

The field of translational research in Duchenne muscular dystrophy (DMD) has been transformed in the last decade by a number of therapeutic targets, mostly studied in ambulant patients. A paucity of studies focus on measures that capture the non-ambulant stage of the disease, and the transition between the ambulant and non-ambulant phase. In this prospective natural history study, we report the results of a comprehensive assessment of respiratory, upper limb function and upper limb muscle strength in a group of 89 DMD boys followed in 3 European countries, 81 receiving corticosteroids, spanning a wide age range (5-18 years) and functional abilities, from ambulant (n = 60) to non-ambulant (n = 29). Respiratory decline could be detected in the early ambulatory phase using Peak Expiratory Flow percentage predicted (PEF%), despite glucocorticoid use (mean annual decline: 4.08, 95% CI [-7.44,-0.72], p = 0.02 in ambulant; 4.81, 95% CI [-6.79,-2.82], p < 0.001 in non-ambulant). FVC% captured disease progression in non-ambulant DMD subjects, with an annual loss of 5.47% (95% CI [-6.48,-4.45], p < 0.001). Upper limb function measured with the Performance of Upper Limb (PUL 1.2) showed an annual loss of 4.13 points (95% CI [-4.79,3.47], p < 0.001) in the non-ambulant cohort. Measures of upper limb strength (MyoGrip and MyoPinch) showed a continuous decline independent of the ambulatory status, when reported as percentage predicted (grip force -5.51%, 95% CI [-6.54,-4.48], p < 0.001 in ambulant and a slower decline -2.86%; 95% CI -3.29,-2.43, p < 0.001, in non-ambulant; pinch force: -2.66%, 95% CI [-3.82,-1.51], p < 0.001 in ambulant and -2.23%, 95% CI [-2.92,-1.53], p < 0.001 in non-ambulant). Furthermore, we also explored the novel concept of a composite endpoint by combining respiratory, upper limb function and force domains: we were able to identify clear clinical progression in patients in whom an isolated measurement of only one of these domains failed to appreciate the yearly change. Our study contributes to the field of natural history of DMD, linking the ambulant and non-ambulant phases of the disease, and suggests that composite scores should be explored further.


Subject(s)
Mobility Limitation , Motor Activity/physiology , Muscle Strength/physiology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Outcome Assessment, Health Care , Respiration Disorders/physiopathology , Upper Extremity/physiopathology , Adolescent , Child , Child, Preschool , Europe , Humans , Male , Muscular Dystrophy, Duchenne/complications , Prospective Studies , Respiration , Respiration Disorders/etiology , Respiratory Function Tests
19.
Hum Mutat ; 29(6): 809-22, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18366090

ABSTRACT

Mutations in the genes encoding collagen VI (COL6A1, COL6A2, and COL6A3) cause Bethlem myopathy (BM) and Ullrich congenital muscular dystrophy (UCMD), two related conditions of differing severity. BM is a relatively mild dominantly inherited disorder characterized by proximal weakness and distal joint contractures. UCMD was originally regarded as an exclusively autosomal recessive condition causing severe muscle weakness with proximal joint contractures and distal hyperlaxity. We and others have subsequently modified this model when we described UCMD patients with heterozygous in-frame deletions acting in a dominant-negative way. Here we report 10 unrelated patients with a UCMD clinical phenotype and de novo dominant negative heterozygous splice mutations in COL6A1, COL6A2, and COL6A3 and contrast our findings with four UCMD patients with recessively acting splice mutations and two BM patients with heterozygous splice mutations. We find that the location of the skipped exon relative to the molecular structure of the collagen chain strongly correlates with the clinical phenotype. Analysis by immunohistochemical staining of muscle biopsies and dermal fibroblast cultures, as well as immunoprecipitation to study protein biosynthesis and assembly, suggests different mechanisms each for exon skipping mutations underlying dominant UCMD, dominant BM, and recessive UCMD. We provide further evidence that de novo dominant mutations in severe UCMD occur relatively frequently in all three collagen VI chains and offer biochemical insight into genotype-phenotype correlations within the collagen VI-related disorders by showing that severity of the phenotype depends on the ability of mutant chains to be incorporated in the multimeric structure of collagen VI.


Subject(s)
Collagen Type VI/genetics , Muscular Dystrophies/genetics , Mutation , RNA Splicing , Cells, Cultured , Collagen Type VI/metabolism , DNA Mutational Analysis , Exons , Fibroblasts/metabolism , Gene Deletion , Humans , Muscle, Skeletal/metabolism , Severity of Illness Index , Skin/cytology
20.
J Neuroimmunol ; 201-202: 6-12, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18707767

ABSTRACT

The Congenital Myasthenic Syndromes (CMS), a group of heterogeneous genetic disorders of neuromuscular transmission, are often misdiagnosed as congenital muscular dystrophy (CMD) or myopathies and present particular management problems. We present our experience of 46 children with CMS, referred to us between 1992-2007 with provisional diagnoses of congenital myopathy (22/46), CMS or limb-girdle myasthenia (9/46), central hypotonia or neurometabolic disease (5/46), myasthenia gravis (4/46), limb-girdle or congenital muscular dystrophy (4/46) and SMA (2/46). Diagnosis was often considerably delayed (up to 18y4 m), despite the early symptoms in most cases. Diagnostic clues in the neonates were feeding difficulties (29/46), hypotonia with or without limb weakness (21/46), ptosis (19/46), respiratory insufficiency (12/46), contractures (4/46) and stridor (6/46). Twenty-five children had delayed motor milestones. Fatigability developed in 43 and a variable degree of ptosis was eventually present in 40. Over the period of the study, the mainstay of EMG diagnosis evolved from repetitive nerve stimulation to stimulation single fibre EMG. The patients were studied by several different operators. 66 EMGs were performed in 40 children, 29 showed a neuromuscular junction abnormality, 7 were myopathic, 2 had possible neurogenic changes and 28 were normal or inconclusive. A repetitive CMAP was detected in only one of seven children with a COLQ mutation and neither of the two children with Slow Channel Syndrome mutations. Mutations have been identified so far in 32/46 children: 10 RAPSN, 7 COLQ, 6 CHRNE, 7 DOK7, 1 CHRNA1 and 1 CHAT. 24 of 25 muscle biopsies showed myopathic changes with fibre size variation; 14 had type-1 fibre predominance. Three cases showed small type-1 fibres resembling fibre type disproportion, and four showed core-like lesions. No specific myopathic features were associated with any of the genes. Twenty children responded to Pyridostigmine treatment alone, 11 to Pyridostigmine with either 3, 4 DAP or Ephedrine and five to Ephedrine alone. Twenty one children required acute or chronic respiratory support, with tracheostomy in 4 and nocturnal or emergency non-invasive ventilation in 9. Eight children had gastrostomy. Another 11 were underweight for height indicative of failure to thrive and required dietetic input. A high index of clinical suspicion, repeat EMG by an experienced electromyographer and, if necessary, a therapeutic trial of Pyridostigmine facilitates the diagnosis of CMS with subsequent molecular genetic confirmation. This guides rational therapy and multidisciplinary management, which may be crucial for survival, particularly in pedigrees where previous deaths have occurred in infancy.


Subject(s)
Myasthenic Syndromes, Congenital/diagnosis , Myasthenic Syndromes, Congenital/therapy , Adolescent , Age of Onset , Biopsy/methods , Child , Child, Preschool , DNA Mutational Analysis , Electromyography , Female , Humans , Infant , Infant, Newborn , Male , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/pathology , Mutation , Myasthenic Syndromes, Congenital/classification , Myasthenic Syndromes, Congenital/physiopathology , Respiration , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL