Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 113
Filter
Add more filters











Publication year range
1.
Neuromuscul Disord ; 31(5): 371-384, 2021 05.
Article in English | MEDLINE | ID: mdl-33685841

ABSTRACT

Our retrospective immunohistochemical study of normal quadriceps muscle biopsies shows that embryonic myosin heavy chains are down-regulated by, or soon after, birth. Fetal myosin heavy chains are down-regulated by 4-6 months. Thus the presence of an appreciable number of fibres with embryonic myosin heavy chains at birth or of fetal myosin heavy chains after 6 months of age suggests a delay in maturation or an underlying abnormality. Regenerating fibres in dystrophic muscle often co-express both embryonic and fetal myosin heavy chains but more fibres with fetal than embryonic myosin heavy chains can occur. Embryonic myosin heavy chains are a useful marker of regeneration but effects of denervation, stress, disuse, and fibre maintenance also have to be taken into account. In neurogenic disorders fibres with embryonic myosin heavy chains are rare but fetal myosin heavy chain expression is common, particularly in 5q spinal muscle atrophy. Nuclear clumps in denervated muscle show fetal and sometimes embryonic myosin heavy chains. Developmentally regulated myosins are useful for highlighting the perifascicular atrophy in juvenile dermatomyositis. Our studies highlight the importance of baseline data for embryonic and fetal myosin heavy chains in human muscle biopsies and the importance of assessing them in a spectrum of neuromuscular disorders.


Subject(s)
Biopsy , Myosin Heavy Chains/metabolism , Quadriceps Muscle/pathology , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Regeneration , Retrospective Studies , Young Adult
2.
Neuropathol Appl Neurobiol ; 43(1): 5-23, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27976420

ABSTRACT

Congenital myopathies are clinically and genetically a heterogeneous group of early onset neuromuscular disorders, characterized by hypotonia and muscle weakness. Clinical severity and age of onset are variable. Many patients are severely affected at birth while others have a milder, moderately progressive or nonprogressive phenotype. Respiratory weakness is a major clinical aspect that requires regular monitoring. Causative mutations in several genes have been identified that are inherited in a dominant, recessive or X-linked manner, or arise de novo. Muscle biopsies show characteristic pathological features such as nemaline rods/bodies, cores, central nuclei or caps. Small type 1 fibres expressing slow myosin are a common feature and may sometimes be the only abnormality. Small cores (minicores) devoid of mitochondria and areas showing variable myofibrillar disruption occur in several neuromuscular disorders including several forms of congenital myopathy. Muscle biopsies can also show more than one structural defect. There is considerable clinical, pathological and genetic overlap with mutations in one gene resulting in more than one pathological feature, and the same pathological feature being associated with defects in more than one gene. Increasing application of whole exome sequencing is broadening the clinical and pathological spectra in congenital myopathies, but pathology still has a role in clarifying the pathogenicity of gene variants as well as directing molecular analysis.


Subject(s)
Myopathies, Structural, Congenital/pathology , Humans , Muscle, Skeletal/pathology
4.
Neuromuscul Disord ; 25(5): 388-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25747004

ABSTRACT

We present follow up data on the original case of 'zebra body myopathy' published by Lake and Wilson in 1975. Pathological features in a second biopsy performed at the age of 29 years included a wide variation in fibre size, multiple split fibres, excess internal nuclei and endomysial connective tissue, rimmed vacuoles, accumulation of myofibrillar material and large 'wiped out' areas lacking stain for oxidative enzymes. The presence of nemaline rods and actin-like filaments in addition to small zebra bodies suggested ACTA1 as a candidate gene. This has been confirmed by the identification of a novel c.1043T.p.Leu348Gln mutation, which probably occurred de novo. This case illustrates that the myopathy associated with zebra bodies is part of the spectrum of myopathies associated with the ACTA1 gene. It also highlights that accumulation of actin filaments is not confined to severe neonatal ACTA1 cases and that progression of weakness can occur in congenital myopathies, as the patient is now wheelchair bound and can only stand with the aid of a walking frame.


Subject(s)
Actins/genetics , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Myopathies, Nemaline/genetics , Adult , Follow-Up Studies , Humans , Male , Muscle, Skeletal/ultrastructure , Muscular Diseases/pathology , Myopathies, Nemaline/pathology
5.
Neuromuscul Disord ; 24(8): 707-12, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24951453

ABSTRACT

The spectrum of RYR1 mutation associated disease encompasses congenital myopathies, exercise induced rhabdomyolysis, malignant hyperthermia susceptibility and King-Denborough syndrome. We report the clinical phenotype of two siblings who presented in infancy with hypotonia and striking fatigable ptosis. Their response to pyridostigimine was striking, but genetic screening for congenital myasthenic syndromes was negative, prompting further evaluation. Muscle MRI was abnormal with a selective pattern of involvement evocative of RYR1-related myopathy. This directed sequencing of the RYR1 gene, which revealed two heterozygous c.6721C>T (p.Arg2241X) nonsense mutations and novel c.8888T>C (p.Leu2963Pro) mutations in both siblings. These cases broaden the RYR1-related disease spectrum to include a myasthenic-like phenotype, including partial response to pyridostigimine. RYR1-related myopathy should be considered in the presence of fatigable weakness especially if muscle imaging demonstrates structural abnormalities. Single fibre electromyography can also be helpful in cases like this.


Subject(s)
Cholinesterase Inhibitors/therapeutic use , Myotonia Congenita/drug therapy , Myotonia Congenita/genetics , Pyridostigmine Bromide/therapeutic use , Ryanodine Receptor Calcium Release Channel/genetics , Biopsy , Codon, Nonsense , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Muscle Fatigue/drug effects , Muscle Fatigue/physiology , Muscles/drug effects , Muscles/pathology , Muscles/physiopathology , Myotonia Congenita/pathology , Myotonia Congenita/physiopathology , Siblings , Treatment Outcome
6.
Neuromuscul Disord ; 23(3): 195-205, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23394784

ABSTRACT

The congenital myopathies are a group of inherited neuromuscular disorders mainly defined on the basis of characteristic histopathological features. We analysed 66 patients assessed at a single centre over a 5 year period. Of the 54 patients where muscle biopsy was available, 29 (54%) had a core myopathy (central core disease, multi-minicore disease), 9 (17%) had nemaline myopathy, 7 (13%) had myotubular/centronuclear myopathy, 2 (4%) had congenital fibre type disproportion, 6 (11%) had isolated type 1 predominance and 1 (2%) had a mixed core-rod myopathy. Of the 44 patients with a genetic diagnosis, RYR1 was mutated in 26 (59%), ACTA1 in 7 (16%), SEPN1 in 7 (16%), MTM1 in 2 (5%), NEB in 1 (2%) and TPM3 in 1 (2%). Clinically, 77% of patients older than 18 months could walk independently. 35% of all patients required ventilatory support and/or enteral feeding. Clinical course was stable or improved in 57/66 (86%) patients, whilst 4 (6%) got worse and 5 (8%) died. These findings indicate that core myopathies are the most common form of congenital myopathies and that more than half can be attributed to RYR1 mutations. The underlying genetic defect remains to be identified in 1/3 of congenital myopathies cases.


Subject(s)
Muscle, Skeletal/pathology , Muscular Diseases/congenital , Muscular Diseases/diagnosis , Adolescent , Age of Onset , Child , Child, Preschool , Disease Progression , Female , Humans , Infant , Infant, Newborn , Male , Muscular Diseases/genetics , Muscular Diseases/pathology , Mutation , United Kingdom
7.
Neuromuscul Disord ; 22(12): 1096-104, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22784669

ABSTRACT

Central Core Disease (CCD) and Multi-minicore Disease (MmD) (the "core myopathies") have been mainly associated with mutations in the skeletal muscle ryanodine receptor (RYR1) and the selenoprotein N (SEPN1) gene. A proportion of cases remain unresolved. Mutations in MYH7 encoding the beta myosin heavy chain protein have been implicated in cardiac and, less frequently, skeletal muscle disorders. Here we report four patients from two families with a histopathological diagnosis of MmD, presenting in childhood with slowly progressive muscle weakness, more proximal in Family 1 and more distal in Family 2, and variable degrees of cardiorespiratory impairment evolving later in life. There was also a strong family history of sudden death in the first family. Muscle biopsies obtained in early childhood showed multiple minicores as the most prominent feature. Sequencing of the MYH7 gene revealed heterozygous missense mutations, c.4399C>G; p.Leu1467Val (exon 32) in Family 1 and c.4763G>C; p.Arg1588Pro (exon 34) in Family 2. These findings suggest MYH7 mutations as another cause of a myopathy with multiple cores, in particular if associated with dominant inheritance and cardiac involvement. However, clinical features previously associated with this genetic background, namely a more distal distribution of weakness and an associated cardiomyopathy, may only evolve over time.


Subject(s)
Cardiac Myosins/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Mutation/genetics , Myopathy, Central Core/genetics , Myosin Heavy Chains/genetics , Adult , Child , Female , Genetic Heterogeneity , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Muscular Diseases/diagnosis , Muscular Diseases/pathology , Myopathy, Central Core/diagnosis , Myopathy, Central Core/pathology , Pedigree , Ryanodine Receptor Calcium Release Channel/genetics
8.
Neuromuscul Disord ; 22(6): 522-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22480491

ABSTRACT

The Dubowitz Neuromuscular Centre is the UK National Commissioning Group referral centre for congenital muscular dystrophy (CMD). This retrospective review reports the diagnostic outcome of 214 UK patients referred to the centre for assessment of 'possible CMD' between 2001 and 2008 with a view to commenting on the variety of disorders seen and the relative frequency of CMD subtypes in this patient population. A genetic diagnosis was reached in 53 of 116 patients fulfilling a strict criteria for the diagnosis of CMD. Within this group the most common diagnoses were collagen VI related disorders (19%), dystroglycanopathy (12%) and merosin deficient congenital muscular dystrophy (10%). Among the patients referred as 'possible CMD' that did not meet our inclusion criteria, congenital myopathies and congenital myasthenic syndromes were the most common diagnoses. In this large study on CMD the diagnostic outcomes compared favourably with other CMD population studies, indicating the importance of an integrated clinical and pathological assessment of this group of patients.


Subject(s)
Muscular Dystrophies/congenital , Muscular Dystrophies/diagnosis , Adolescent , Adult , Child , Child, Preschool , Diagnostic Services , Female , Genetic Testing , Humans , Infant , Male , Muscular Dystrophies/genetics , Retrospective Studies , United Kingdom
10.
Neuromuscul Disord ; 22(2): 139-48, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22075033

ABSTRACT

Mutations in COL6A1, COL6A2 and COL6A3 genes result in collagen VI myopathies: Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM) and intermediate phenotypes. At present, none of the existing diagnostic techniques for evaluating collagen VI expression is quantitative, and the detection of subtle changes in collagen VI expression remains challenging. We investigated flow cytometry analysis as a means of quantitatively measuring collagen VI in primary fibroblasts and compared this method with the standard method of fibroblast collagen VI immunohistochemical analysis. Eight UCMD and five BM molecularly confirmed patients were studied and compared to five controls. Flow cytometry analysis consistently detected a reduction of collagen VI of at least 60% in all UCMD cases. In BM cases the levels of collagen VI were variable but on average 20% less than controls. Flow cytometry analysis provides an alternative method for screening for collagen VI deficiency at the protein level in a quantitative, time and cost-effective manner.


Subject(s)
Collagen Type VI/deficiency , Flow Cytometry , Muscular Dystrophies/diagnosis , Adolescent , Adult , Child , Collagen Type VI/genetics , Collagen Type VI/metabolism , Fibroblasts/metabolism , Humans , Middle Aged , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Mutation/genetics , Young Adult
11.
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
12.
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
13.
Neurology ; 71(22): 1757-63, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18633133

ABSTRACT

OBJECTIVES: Spinal muscular atrophy (SMA) is an autosomal recessive disorder characterized by loss of lower motor neurons during early or postnatal development. Severity is variable and is inversely related to the levels of survival of motor neurons (SMN) protein. The aim of this study was to produce a two-site ELISA capable of measuring both the low, basal levels of SMN protein in cell cultures from patients with severe SMA and small increases in these levels after treatment of cells with drugs. METHODS: A monoclonal antibody against recombinant SMN, MANSMA1, was selected for capture of SMN onto microtiter plates. A selected rabbit antiserum against refolded recombinant SMN was used for detection of the captured SMN. RESULTS: The ratio of SMN levels in control fibroblasts to levels in SMA fibroblasts was greater than 3.0, consistent with Western blot data. The limit of detection was 0.13 ng/mL and SMN could be measured in human NT-2 neuronal precursor cells grown in 96-well culture plates (3 x 10(4) cells per well). Increases in SMN levels of 50% were demonstrable by ELISA after 24 hours treatment of 10(5) SMA fibroblasts with valproate or phenylbutyrate. CONCLUSION: A rapid and specific two-site, 96-well ELISA assay, available in kit format, can now quantify the effects of drugs on survival of motor neurons protein levels in cell cultures.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System Agents/therapeutic use , Enzyme-Linked Immunosorbent Assay , Motor Neurons , Muscular Atrophy, Spinal/genetics , Survival of Motor Neuron 1 Protein/genetics , Blotting, Western , Cell Line , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay/methods , Fibroblasts/drug effects , Humans , Motor Neurons/drug effects , Muscular Atrophy, Spinal/blood , Muscular Atrophy, Spinal/physiopathology , Phenylbutyrates/pharmacology , Phenylbutyrates/therapeutic use , Predictive Value of Tests , Recombinant Proteins/genetics , Survival of Motor Neuron 1 Protein/blood , Up-Regulation/drug effects , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
14.
Neuromuscul Disord ; 17(2): 174-9, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17236770

ABSTRACT

Severe diaphragmatic weakness in infancy is rare. Common causes include structural myopathies, neuromuscular transmission defects, or anterior horn cell dysfunction (spinal muscular atrophy with respiratory distress, SMARD1). We describe a form of infantile diaphragmatic weakness without mutations in the SMARD1 gene, in which pathological and clinical features differ from known conditions, and investigations suggest a myopathy. We identified seven cases in four families. All presented soon after birth with feeding and breathing difficulties, marked head lag, facial weakness, and preserved antigravity movements in the limbs, with arms weaker than legs. All had paradoxical breathing and paralysis of at least one hemi-diaphragm. All required gastrostomy feeding, and all became ventilator-dependent. Investigations included myopathic EMG, muscle biopsy showing myopathic changes, normal electrophysiology and no mutations in SMN1 or IGHMBP2. These seven infants are affected by a myopathic condition clinically resembling SMARD1. However, its pathogenesis appears to be a myopathy affecting predominantly the diaphragm.


Subject(s)
DNA-Binding Proteins/genetics , Diaphragm/physiopathology , Muscle Weakness/congenital , Muscle Weakness/genetics , Muscular Diseases/congenital , Muscular Diseases/genetics , Transcription Factors/genetics , Creatine Kinase/metabolism , Electromyography , Enteral Nutrition , Extremities/physiopathology , Facial Muscles/physiopathology , Female , Growth/physiology , Humans , Infant , Infant, Newborn , Movement/physiology , Muscle Weakness/physiopathology , Muscular Diseases/physiopathology , Respiration, Artificial , Respiratory Mechanics/physiology
15.
Acta Myol ; 26(3): 129-35, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18646561

ABSTRACT

Muscular dystrophies are a clinically and genetically heterogeneous group of disorders. Until recently most of the proteins associated with muscular dystrophies were believed to be proteins of the sarcolemma associated with reinforcing the plasma membrane or in facilitating its re-sealing following injury. In the last few years a novel and frequent pathogenic mechanism has been identified that involves the abnormal glycosylation of alpha-dystroglycan (ADG). This peripheral membrane protein undergoes complex and crucial glycosylation steps that enable it to interact with LG domain containing extracellular matrix proteins such as laminins, agrin and perlecan. Mutations in six genes (POMT1, POMT2, POMGnT1, fukutin, FKRP and LARGE) have been identified in patients with reduced glycosylation of ADG. While initially a clear correlation between gene defect and phenotype was observed for each of these 6 genes (for example, Walker Warburg syndrome was associated with mutations in POMT1 and POMT2, Fukuyama congenital muscular dystrophy associated with fukutin mutations, and Muscle Eye Brain disease associated with POMGnT1 mutations), we have recently demonstrated that allelic mutations in each of these 6 genes can result in a much wider spectrum of clinical conditions. Thus, the crucial aspect in determining the phenotypic severity is not which gene is primarily mutated, but how severely the mutation affects the glycosylation of ADG. Systematic mutation analysis of these 6 glycosyltransferases in patients with a dystroglycan glycosylation disorder identifies mutations in approximately 65% suggesting that more genes have yet to be identified.


Subject(s)
Dystroglycans/metabolism , Muscular Dystrophies/metabolism , DNA/genetics , Dystroglycans/genetics , Glycosylation , Humans , Muscular Dystrophies/genetics , Mutation
16.
Neuromuscul Disord ; 16(9-10): 571-82, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16935502

ABSTRACT

Ullrich congenital muscular dystrophy (UCMD) is caused by recessive and dominant mutations in COL6A genes. We have analysed collagen VI expression in 14 UCMD patients. Sequencing of COL6A genes had identified homozygous and heterozygous mutations in 12 cases. Analysis of collagen VI in fibroblast cultures derived from eight of these patients showed reduced extracellular deposition in all cases and intracellular collagen VI staining in seven cases. This was observed even in cases that showed normal collagen VI labelling in skin biopsies. Collagen VI immunolabelling was reduced in all the available muscle biopsies. When comparisons were possible no correlation was seen between the extent of the reduction in the muscle and fibroblast cultures, the mode of inheritance or the severity of the clinical phenotype. Mutations affecting glycine substitutions in the conserved triple helical domain were common and all resulted in reduced collagen VI. This study expands the spectrum of collagen VI defects and shows that analysis of skin fibroblasts may be a useful technique for the detection of collagen VI abnormalities. In contrast, immunohistochemical analysis of skin biopsies may not always reveal an underlying collagen VI defect.


Subject(s)
Collagen Type VI/genetics , Fibroblasts/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Skin/metabolism , Adolescent , Adult , Cells, Cultured , Child , Collagen Type VI/biosynthesis , DNA Mutational Analysis , Disease Progression , Fibroblasts/pathology , Genes, Dominant/genetics , Genes, Recessive/genetics , Genetic Markers/genetics , Genetic Predisposition to Disease/genetics , Genetic Testing , Humans , Immunohistochemistry/standards , Inheritance Patterns/genetics , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Dystrophies/physiopathology , Mutation/genetics , Phenotype , Predictive Value of Tests , Skin/pathology , Skin/physiopathology
17.
Brain ; 129(Pt 5): 1260-8, 2006 May.
Article in English | MEDLINE | ID: mdl-16585054

ABSTRACT

Individuals with the same genetic disorder often show remarkable differences in clinical severity, a finding generally attributed to the genetic background. We identified two patients with genetically proven Emery-Dreifuss muscular dystrophy (EDMD) who followed an unusual course and had uncommon clinicopathological findings. We hypothesized digenic inheritance and looked for additional molecular explanations. Mutations in additional separate genes were identified in both patients. The first patient was a member of a family with molecularly proven X-linked EDMD. However, the clinical features were unusually severe for this condition in the propositus: he presented at 2.5 years with severe proximal weakness and markedly elevated serum creatine kinase. Muscle weakness rapidly progressed, leading to loss of independent ambulation by the age of 12. In addition, the patient developed cardiac conduction system disease requiring pacing at the age of 11 and severe dilated cardiomyopathy in the early teens. Despite pacing, he had several syncopal episodes attributed to ventricular dysrhythmias. As these resemble the cardiac features of patients with the autosomal dominant variant of EDMD, we examined the lamin A/C gene, identifying a de-novo mutation in the propositus. The second patient had a cardioskeletal myopathy, similar to his mother who had died more than 20 years previously. Because of the dominant family history, a laminopathy was suspected and a mutation in exon 11 of the LMNA gene was identified. This mutation, however, was not present in his mother, but instead, surprisingly, was identified in his virtually asymptomatic father. Unusual accumulations of desmin found in the cardiac muscle of the propositus prompted us to examine the desmin gene in this patient, and in so doing, we identified a desmin mutation, in addition to the LMNA mutation in the propositus. These cases suggest that separate mutations in related proteins that are believed to interact, or that represent different parts of a presumed functional pathway, may synergistically contribute to disease severity in autosomal dominant EDMD. Furthermore, digenic inheritance may well contribute to the clinical severity of many other neuromuscular disorders.


Subject(s)
Desmin/genetics , Membrane Proteins/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Thymopoietins/genetics , Adolescent , Adult , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Child, Preschool , Desmin/metabolism , Female , Humans , Laminin/genetics , Laminin/metabolism , Magnetic Resonance Imaging , Male , Membrane Proteins/metabolism , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Emery-Dreifuss/pathology , Myocardium/metabolism , Myocardium/ultrastructure , Nuclear Proteins , Pedigree , Thymopoietins/metabolism
18.
Neurology ; 65(12): 1930-5, 2005 Dec 27.
Article in English | MEDLINE | ID: mdl-16380615

ABSTRACT

BACKGROUND: Minicore myopathy (multi-minicore disease [MmD]) is a congenital myopathy characterized by multifocal areas with loss of oxidative activity on muscle biopsy. MmD is clinically heterogeneous and distinct phenotypes have been associated with recessive mutations in either the selenoprotein N (SEPN1) or the skeletal muscle ryanodine receptor (RYR1) gene, also implicated in central core disease and malignant hyperthermia. External ophthalmoplegia is an additional finding in a subset of patients with MmD. OBJECTIVE: To clinically and genetically examine families with MmD and external ophthalmoplegia. METHODS: The authors investigated 11 affected individuals from 5 unrelated families. Clinical, histopathologic, and imaging studies were performed and RYR1 haplotyping and mutational analysis were carried out. RESULTS: All patients had multiple cores involving the entire fiber diameter on longitudinal sections. Weakness and wasting in the shoulder girdle, scoliosis, moderate respiratory impairment, and feeding difficulties were prominent. In contrast to SEPN1-related myopathies, soleus was more severely affected than gastrocnemius on muscle MRI. Haplotyping suggested linkage to the RYR1 locus in informative families and mutational screening revealed four novel RYR1 mutations in three unrelated families; in addition, functional haploinsufficiency was found in one allele of two recessive cases. CONCLUSION: These findings expand the phenotypic spectrum associated with mutations in the skeletal muscle ryanodine receptor (RYR1) gene. Recessive mutations of domains commonly affected in malignant hyperthermia appear to be particularly prevalent in multi-minicore disease with external ophthalmoplegia and might suggest a different pathomechanism from that involved in central core disease.


Subject(s)
Genetic Predisposition to Disease/genetics , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Mutation/genetics , Ophthalmoplegia/genetics , Ryanodine Receptor Calcium Release Channel/genetics , Adolescent , Adult , Biopsy , Child , Chromosomes, Human, Pair 19/genetics , DNA Mutational Analysis , Genetic Markers , Genetic Testing , Haplotypes , Humans , Ligaments/pathology , Ligaments/physiopathology , Middle Aged , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Muscular Diseases/pathology , Muscular Diseases/physiopathology , Oculomotor Muscles/metabolism , Oculomotor Muscles/pathology , Oculomotor Muscles/physiopathology , Ophthalmoplegia/pathology , Ophthalmoplegia/physiopathology , Pedigree , Syndrome
19.
Neuromuscul Disord ; 15(11): 779-85, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16198105

ABSTRACT

Utrophin is an autosomal homologue of dystrophin, abnormal expression of which is responsible for X-linked Duchenne and Becker muscular dystrophy. In normal mature muscle utrophin is confined to blood vessels, nerves and myotendinous and neuromuscular junctions. When dystrophin is absent utrophin is abundant on the sarcolemma. This has raised the possibility that up-regulation of utrophin may be of therapeutic benefit. Two full-length transcripts of utrophin, A and B, have been identified, which are regulated by alternatively spliced 5' promoters. In dystrophic mouse muscle, the A isoform is present on the sarcolemma, whereas the B form is confined to blood vessels. We show here using immunohistochemistry and human isoform-specific antibodies that A- and B-utrophin localisation is the same in human muscle. The A isoform is present on the sarcolemma of foetal human muscle fibres, regenerating fibres, fibres deficient in dystrophin and on blood vessels and neuromuscular junctions. B-utrophin is only detected on blood vessels. We also show that muscle adjacent to some soft tissue tumours shows increased sarcolemmal utrophin-A, showing that utrophin and dystrophin can simultaneously localise to the sarcolemma and raising the possibility that factor(s) from the tumour cells or accompanying inflammatory cells may have a role in regulating utrophin.


Subject(s)
Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Neoplasms/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Adolescent , Dystrophin/metabolism , Embryo, Mammalian , Humans , Immunohistochemistry/methods , Infant , Intercellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Protein Isoforms/metabolism , Proteins/metabolism , Utrophin/classification
20.
Cell Mol Life Sci ; 62(7-8): 809-23, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15868406

ABSTRACT

The congenital muscular dystrophies are a clinically and genetically heterogeneous group of neuromuscular disorders. Each form has a characteristic phenotype, but there is overlap between some entities and their classification is based on a combination of clinical features and the primary or secondary protein defect. Recent studies have identified the genetic basis of a number of congenital muscular dystrophies (11 genes in total) and have recognised a novel pathological mechanism that highlights the importance of the correct posttranslational processing of proteins, in particular alpha-dystroglycan. Diagnosis of these conditions has been aided by the availability of specific antibodies for each protein and a better understanding of the protein changes that accompany each condition. In this review we present the major molecular, clinical and diagnostic aspects of each group of congenital muscular dystrophy with an emphasis in the more recent developments.


Subject(s)
Dystroglycans/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophies/metabolism , Antigens, CD/metabolism , Humans , Integrin alpha Chains/metabolism , Laminin/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophies/congenital , Muscular Dystrophies/genetics , Muscular Dystrophies/physiopathology , Protein Processing, Post-Translational
SELECTION OF CITATIONS
SEARCH DETAIL