Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 46
Filtrer
1.
J Neuromuscul Dis ; 7(4): 523-534, 2020.
Article de Anglais | MEDLINE | ID: mdl-32538864

RÉSUMÉ

Spinal muscular atrophy (SMA) is a progressive autosomal recessive motor neuron disease which affects 1 in 6,000-10,000 live births, caused by loss of the survival motor neuron 1 gene (SMN1). A major focus of therapeutic developments has been on increasing the full-length SMN protein by increasing the inclusion of exon 7 in SMN2 transcripts, enhancing SMN2 gene expression, stabilizing the SMN protein or replacing the SMN1 gene.In June 2017, FDA and EMA have approved the antisense oligonucleotide Nusinersen as the first treatment for all SMA subtypes without age restriction. While prominent treatment effects have been observed in the earlier stages of the disease and in patients up to 15 years of age, there is only limited data from clinical trials in adult SMA patients. First real-world data from neuromuscular clinical centers suggest a therapeutic benefit of nusinersen with a favourable safety profile also in adult SMA patients: in several cases, relevant improvements of motor function is achieved, which might lead to enhanced autonomy in daily life activities and improved quality of life. Systematic follow-up of the motor status with validated instruments is crucial for an adequate monitoring of the therapeutic effects but most of the widely used scales and scores have been developed and evaluated for the pediatric population only. International neuromuscular experts have met in Frankfurt/Main, Germany in May 2019 to discuss relevant aspects of the diagnostic pathway and patient management in adult SMA. The recommendations and challenges in this patient population are discussed.


Sujet(s)
Amyotrophie spinale/diagnostic , Amyotrophie spinale/traitement médicamenteux , Oligonucléotides/usage thérapeutique , /normes , Guides de bonnes pratiques cliniques comme sujet/normes , Adulte , Congrès comme sujet , Humains , /méthodes
2.
Eur J Neurol ; 27(8): 1390-1396, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32400062

RÉSUMÉ

BACKGROUND AND PURPOSE: Systematic research on the effect of Charcot-Marie-Tooth (CMT) disease on the outcome of pregnancy and conversely the effect of pregnancy on neuropathy is still sparse. METHODS: A clinical cohort study and cross-sectional study within the German CMT-NET was conducted between 2016 and 2019. Inclusion criteria were a confirmed diagnosis of CMT and at least one completed pregnancy after 1990. All participants agreed to fill in questionnaires and have their medical files reviewed. RESULTS: The study group comprised 54 women with a total of 98 pregnancies. The mean age at onset of CMT disease was 12.6 years (range 0-37 years). Fifty (92%) patients had autosomal dominant CMT; two patients each (4%) had X-linked and autosomal recessive CMT. Forty patients (74%) had a PMP22 gene duplication (CMT1A). Obstetric complications did not differ significantly from a German reference population, neither in the whole group nor in the CMT1A group. Overall there was no increased newborn morbidity and mortality. About one-third of patients reported exacerbation of CMT disease in or after pregnancy. No adverse effects of anaesthesia were reported. Most participants stressed a positive attitude and awareness of challenges associated with pregnancy. Important issues were assistance and support in caring for the family. DISCUSSION: In line with findings from our previous study undertaken in the 1990s, there were no increased complication rates for pregnancy and delivery. These results are reassuring for the vast majority of CMT patients and are important for family planning and clinical care.


Sujet(s)
Maladie de Charcot-Marie-Tooth , Adolescent , Adulte , Maladie de Charcot-Marie-Tooth/épidémiologie , Maladie de Charcot-Marie-Tooth/génétique , Enfant , Enfant d'âge préscolaire , Études de cohortes , Études transversales , Femelle , Allemagne/épidémiologie , Humains , Nourrisson , Nouveau-né , Grossesse , Issue de la grossesse/épidémiologie , Jeune adulte
3.
Nat Med ; 26(2): 207-214, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31988462

RÉSUMÉ

Frameshift mutations in the DMD gene, encoding dystrophin, cause Duchenne muscular dystrophy (DMD), leading to terminal muscle and heart failure in patients. Somatic gene editing by sequence-specific nucleases offers new options for restoring the DMD reading frame, resulting in expression of a shortened but largely functional dystrophin protein. Here, we validated this approach in a pig model of DMD lacking exon 52 of DMD (DMDΔ52), as well as in a corresponding patient-derived induced pluripotent stem cell model. In DMDΔ52 pigs1, intramuscular injection of adeno-associated viral vectors of serotype 9 carrying an intein-split Cas9 (ref. 2) and a pair of guide RNAs targeting sequences flanking exon 51 (AAV9-Cas9-gE51) induced expression of a shortened dystrophin (DMDΔ51-52) and improved skeletal muscle function. Moreover, systemic application of AAV9-Cas9-gE51 led to widespread dystrophin expression in muscle, including diaphragm and heart, prolonging survival and reducing arrhythmogenic vulnerability. Similarly, in induced pluripotent stem cell-derived myoblasts and cardiomyocytes of a patient lacking DMDΔ52, AAV6-Cas9-g51-mediated excision of exon 51 restored dystrophin expression and amelioreate skeletal myotube formation as well as abnormal cardiomyocyte Ca2+ handling and arrhythmogenic susceptibility. The ability of Cas9-mediated exon excision to improve DMD pathology in these translational models paves the way for new treatment approaches in patients with this devastating disease.


Sujet(s)
Dystrophine/génétique , Mutation avec décalage du cadre de lecture , Édition de gène/méthodes , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , /génétique , Animaux , Modèles animaux de maladie humaine , Exons , Femelle , Régulation de l'expression des gènes , Thérapie génétique , Génome , Défaillance cardiaque/génétique , Défaillance cardiaque/thérapie , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Mâle , Spectrométrie de masse , Muscles squelettiques/métabolisme , Muscles/métabolisme , Myoblastes/métabolisme , Myocytes cardiaques/métabolisme , Protéome , Suidae
4.
J Neuromuscul Dis ; 4(4): 315-325, 2017.
Article de Anglais | MEDLINE | ID: mdl-29172004

RÉSUMÉ

BACKGROUND: Early-onset myopathies are a heterogeneous group of neuromuscular diseases with broad clinical, genetic and histopathological overlap. The diagnostic approach has considerably changed since high throughput genetic methods (next generation sequencing, NGS) became available. OBJECTIVE: We present diagnostic subgroups in a single neuromuscular referral center and describe an algorithm for the diagnostic work-up. METHODS: The diagnostic approach of 98 index patients was retrospectively analysed. In 56 cases targeted sequencing of a known gene was performed, in 44 patients NGS was performed using large muscle specific panels, and in 12 individuals whole exome sequencing (WES) was undertaken. One patient was diagnosed via array CGH. Clinical features of all patients are provided. RESULTS: The final diagnosis could be found in 63 out of 98 patients (64%) with molecular genetic analysis. In 55% targeted gene sequencing could establish the genetic diagnosis. However, this rate largely depended on the presence of distinct histological or clinical features. NGS (large myopathy-related panels and WES) revealed genetic diagnosis in 58.5% (52% and 67%, respectively). The genes detected by WES in our cohort of patients were all covered by the panels. Based on our findings we propose an algorithm for a practical diagnostic approach.Prevalences:MTM1- and LAMA2-patients are the two biggest subgroups, followed by SEPN1-, RYR1- and Collagen VI-related diseases. 31% of genetically confirmed cases represents a group with overlap between "congenital myopathies (CM)" and "congenital muscular dystrophies (CMD)". In 36% of the patients a specific genetic diagnosis could not be assigned. CONCLUSIONS: A final diagnosis can be confirmed by high throughput genetic analysis in 58.5% of the cases, which is a higher rate than reported in the literature for muscle biopsy and should in many cases be considered as a first diagnostic tool. NGS cannot replace neuromuscular expertise and a close discussion with the geneticists on NGS is mandatory. Targeted candidate gene sequencing still plays a role in selected cases with highly suspicious clinical or histological features. There is a relevant clinical and genetic overlap between the entities CM and CMD.


Sujet(s)
Maladies musculaires/diagnostic , Maladies musculaires/épidémiologie , Âge de début , Algorithmes , Allemagne , Humains , Maladies musculaires/génétique , Prévalence , Études rétrospectives , Analyse de séquence
5.
Nervenarzt ; 88(7): 736-743, 2017 Jul.
Article de Allemand | MEDLINE | ID: mdl-28573363

RÉSUMÉ

Myopathies and mitochondrial diseases pose a major challenge in diagnosis due to the multitude of different entities and - in the case of mitochondriopathies - the possible involvement of multiple organs. Furthermore, there is broad clinical variability within particular diseases; patients with hereditary myopathy, for example, can show great phenotypic variability despite identical genetic defects. In addition to environmental factors, gender-specific influences, and the degree of heteroplasmy in mitochondrial diseases, the existence of disease-modifying genes has long been assumed as an explanation. In recent years, risk genes, which can influence the course of disease, have been identified for some myopathies and mitochondrial diseases. The precise role of these disease-modifying genes in the pathogenesis of the diseases is largely unexplained and requires further research.


Sujet(s)
Maladies mitochondriales/génétique , Maladies musculaires/génétique , Diagnostic différentiel , Ataxie de Friedreich/diagnostic , Ataxie de Friedreich/génétique , Prédisposition génétique à une maladie/génétique , Génotype , Humains , Maladies mitochondriales/diagnostic , Maladies musculaires/diagnostic , Dystrophies musculaires/diagnostic , Dystrophies musculaires/génétique , Atrophie optique héréditaire de Leber/diagnostic , Atrophie optique héréditaire de Leber/génétique , Phénotype
6.
Neurol Genet ; 2(4): e89, 2016 Aug.
Article de Anglais | MEDLINE | ID: mdl-27602406

RÉSUMÉ

OBJECTIVE: To describe the baseline clinical and functional characteristics of an international cohort of 193 patients with dysferlinopathy. METHODS: The Clinical Outcome Study for dysferlinopathy (COS) is an international multicenter study of this disease, evaluating patients with genetically confirmed dysferlinopathy over 3 years. We present a cross-sectional analysis of 193 patients derived from their baseline clinical and functional assessments. RESULTS: There is a high degree of variability in disease onset, pattern of weakness, and rate of progression. No factor, such as mutation class, protein expression, or age at onset, accounted for this variability. Among patients with clinical diagnoses of Miyoshi myopathy or limb-girdle muscular dystrophy, clinical presentation and examination was not strikingly different. Respiratory impairment and cardiac dysfunction were observed in a minority of patients. A substantial delay in diagnosis was previously common but has been steadily reducing, suggesting increasing awareness of dysferlinopathies. CONCLUSIONS: These findings highlight crucial issues to be addressed for both optimizing clinical care and planning therapeutic trials in dysferlinopathy. This ongoing longitudinal study will provide an opportunity to further understand patterns and variability in disease progression and form the basis for trial design.

10.
Brain ; 132(Pt 1): 147-55, 2009 Jan.
Article de Anglais | MEDLINE | ID: mdl-19015158

RÉSUMÉ

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.


Sujet(s)
Ciclosporine/pharmacologie , Mitochondries/physiologie , Dystrophies musculaires/anatomopathologie , Adolescent , Cellules cultivées , Enfant , Enfant d'âge préscolaire , Collagène de type VI/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Humains , Nourrisson , Nouveau-né , Mâle , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries du muscle/effets des médicaments et des substances chimiques , Mitochondries du muscle/physiologie , Protéines de transport de la membrane mitochondriale/antagonistes et inhibiteurs , Protéines de transport de la membrane mitochondriale/métabolisme , Pore de transition de perméabilité mitochondriale , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Dystrophies musculaires/métabolisme , Myoblastes squelettiques/effets des médicaments et des substances chimiques , Myoblastes squelettiques/métabolisme , Rhodamines , Peau/métabolisme , Jeune adulte
11.
Eur J Neurol ; 15(5): 525-9, 2008 May.
Article de Anglais | MEDLINE | ID: mdl-18355305

RÉSUMÉ

Mutations in the epsilon-sarcoglycan gene (SGCE) can cause autosomal dominant inherited myoclonus-dystonia (M-D). Defects in other sarcoglycans; alpha-, beta-, gamma-, and delta can cause autosomal recessive inherited limb girdle muscular dystrophies. epsilon- and alpha-sarcoglycans are very homologous and may substitute for one-another in different tissues. We therefore investigated whether mutations in SGCE also cause abnormalities of skeletal and myocardial muscle. Six patients with clinically and genetically verified M-D and no signs of limb-girdle muscular dystrophy were included. Skeletal muscle biopsies were obtained from all patients, and endomyocardial muscle biopsy from one of the patients. Morphological and immunohistological investigations were performed and compared with controls. Histological and immunohistological investigations of muscle and clinical assessment of muscle strength and mass showed no difference between M-D patients and controls. Our findings indicate that patients with M-D have no signs or symptoms of muscle disease. This suggests a different role of the sarcoglycan complex epsilonbetagammadelta versus alphabetagammadelta complex in humans, as earlier suggested in rodents.


Sujet(s)
Dystonie/anatomopathologie , Muscles squelettiques/anatomopathologie , Myocarde/anatomopathologie , Myoclonie/anatomopathologie , Sarcoglycanes/génétique , Adulte , Biopsie/méthodes , Creatine kinase/sang , Dystonie/génétique , Femelle , Humains , Mâle , Muscles squelettiques/métabolisme , Mutation/génétique , Myocarde/métabolisme , Myoclonie/génétique
13.
Acta Myol ; 27: 54-8, 2008 Oct.
Article de Anglais | MEDLINE | ID: mdl-19364062

RÉSUMÉ

Calpainopathy is an autosomal-recessive limb girdle muscular dystrophy (LGMD2A) characterized by selective atrophy and weakness of proximal limb girdle muscles. The clinical phenotype of the disease is highly variable inter-familial, but little is known about intra-familial variability. This study reports the phenotypic variability in eight sibling pairs with genetically proven LGMD2A. Although siblings with identical mutations were often similarly affected, in some families the age of onset and the clinical course varied considerably.


Sujet(s)
Dystrophies musculaires des ceintures/génétique , Phénotype , Adolescent , Adulte , Calpain/génétique , Enfant , Femelle , Humains , Mâle , Protéines du muscle/génétique , Études rétrospectives , Fratrie , Jeune adulte
14.
Clin Neuropathol ; 26(5): 232-40, 2007.
Article de Anglais | MEDLINE | ID: mdl-17907600

RÉSUMÉ

Inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD, MIM 167320) is a recently identified autosomal dominant disorder due to mutations in the valosin-containing protein (VCP) that affects muscle, bone and brain. Brain involvement and neuropsychological findings of IBMPFD have not been described in detail. A patient carried a novel heterozygous base pair change, 47832C>T, in the VCP gene that resulted in substitution of an arginine residue by cysteine at position 93 (R93C). He presented first with myopathy while bone involvement remained subclinical. The patient developed behavioral abnormalities in his 60s and showed frank personality change with fluent empty speech at the age of 74 years. This syndrome was best classified as semantic dementia. Magnetic resonance imaging disclosed slight but progressive cerebral atrophy with prominent callosal and frontal white matter loss. Positron emission tomography demonstrated glucose hypometabolism of the frontal and temporal lobes disproportionate to their structural involvement. This first comprehensive clinical and neuroimaging study in IBMPFD may raise the awareness among clinicians as well as basic scientists for this exemplary genetic model of dementia.


Sujet(s)
Adenosine triphosphatases/génétique , Encéphale/anatomopathologie , Protéines du cycle cellulaire/génétique , Démence/génétique , Démence/anatomopathologie , Maladies neurodégénératives/génétique , Maladies neurodégénératives/anatomopathologie , Sujet âgé , Analyse de mutations d'ADN , Démence/physiopathologie , Prédisposition génétique à une maladie , Humains , Imagerie par résonance magnétique , Mâle , Muscles squelettiques/anatomopathologie , Mutation , Myosite à inclusions/génétique , Myosite à inclusions/anatomopathologie , Myosite à inclusions/physiopathologie , Maladies neurodégénératives/physiopathologie , Tests neuropsychologiques , Maladie de Paget des os/génétique , Maladie de Paget des os/anatomopathologie , Maladie de Paget des os/physiopathologie , Réaction de polymérisation en chaîne , Tomographie par émission de positons , Protéine contenant la valosine
15.
Neurology ; 69(7): 655-9, 2007 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-17698786

RÉSUMÉ

Mutations in GNE encoding UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) cause hereditary inclusion body myopathy (HIBM). To define the role of GNE mutations in HIBM pathogenesis, GNE protein expression was analyzed. GNE protein is expressed at equal levels in HIBM patients and normal control subjects. Immunofluorescence detection of GNE did not reveal any mislocalization of GNE in skeletal muscle. We conclude that impaired GNE function, not lack of expression, may be the key pathogenic factor in HIBM. For diagnostic purposes, direct genetic analysis of the GNE gene in patients with IBM will remain the mainstay and is not aided by immunohistochemistry or immunoblotting using antibodies against the GNE protein.


Sujet(s)
Régulation de l'expression des gènes codant pour des enzymes , Complexes multienzymatiques/biosynthèse , Complexes multienzymatiques/génétique , Myosite à inclusions/enzymologie , Myosite à inclusions/génétique , Adulte , Carbohydrate epimerases/biosynthèse , Carbohydrate epimerases/génétique , Lignée cellulaire , Femelle , Humains , Mâle , Mutation , Phosphotransferases (Alcohol Group Acceptor)/biosynthèse , Phosphotransferases (Alcohol Group Acceptor)/génétique , Fractions subcellulaires/enzymologie
16.
Brain ; 130(Pt 6): 1485-96, 2007 Jun.
Article de Anglais | MEDLINE | ID: mdl-17439987

RÉSUMÉ

In 1965, an adult-onset, autosomal dominant disorder with a peculiar scapuloperoneal distribution of weakness and atrophy was described in a large, multi-generation kindred and named 'scapuloperoneal syndrome type Kaeser' (OMIM #181400). By genetic analysis of the original kindred, we discovered a heterozygous missense mutation of the desmin gene (R350P) cosegregating with the disorder. Moreover, we detected DES R350P in four unrelated German families allowing for genotype-phenotype correlations in a total of 15 patients carrying the same mutation. Large clinical variability was recognized, even within the same family, ranging from scapuloperoneal (n = 2, 12%), limb girdle (n = 10, 60%) and distal phenotypes (n = 3, 18%) with variable cardiac (n = 7, 41%) or respiratory involvement (n = 7, 41%). Facial weakness, dysphagia and gynaecomastia were frequent additional symptoms. Overall and within each family, affected men seemingly bear a higher risk of sudden, cardiac death as compared to affected women. Moreover, histological and immunohistochemical examination of muscle biopsy specimens revealed a wide spectrum of findings ranging from near normal or unspecific pathology to typical, myofibrillar changes with accumulation of desmin. This study reveals that the clinical and pathological variability generally observed in desminopathies may not be attributed to the nature of the DES mutation alone, but may be influenced by additional genetic and epigenetic factors such as gender. In addition, mutations of the desmin gene should be considered early in the diagnostic work-up of any adult-onset, dominant myopathy, even if specific myofibrillar pathology is absent.


Sujet(s)
Desmine/génétique , Myopathies distales/génétique , Mutation faux-sens , Adulte , Sujet âgé de 80 ans ou plus , Biopsie , Analyse de mutations d'ADN/méthodes , Myopathies distales/anatomopathologie , Femelle , Haplotypes , Humains , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Fibres musculaires squelettiques/anatomopathologie , Muscles squelettiques/anatomopathologie , Dystrophies musculaires des ceintures/génétique , Dystrophies musculaires des ceintures/anatomopathologie , Pedigree , Phénotype , Facteurs sexuels , Syndrome
17.
Radiology ; 242(1): 85-93, 2007 Jan.
Article de Anglais | MEDLINE | ID: mdl-17090714

RÉSUMÉ

PURPOSE: To prospectively evaluate the cost and effectiveness of magnetic resonance (MR) imaging performed to exclude the need for arthroscopy in patients with nonacute knee symptoms who are highly suspected clinically of having intraarticular knee abnormality. MATERIALS AND METHODS: The study was approved by the institutional review boards of three hospitals; informed patient consent was obtained. All 584 included patients (406 male, 178 female; mean age, 31.1 years+/-8.0 [standard deviation]) underwent MR imaging. Patients with an MR result positive for the diagnosis of intraarticular knee abnormality underwent arthroscopy (group A). Patients with a negative MR result were randomly assigned to undergo either conservative (group B) or arthroscopic (group C) treatment. Treatment was considered effective if the Noyes function score had increased 10% or more at 6 months. A cost analysis was performed from a societal perspective to compare the treatment strategy involving MR imaging with the strategy not involving MR imaging. RESULTS: Of the 584 patients, 294 (50.3%) were assigned to group A; 149 (25.5%), to group B; and 141 (24.1%), to group C. At 6 months, the number of patients effectively treated in group B (conservative treatment) was a mean of 5.1%+/-10.0 larger than the number of patients effectively treated in group C (arthroscopy). Owing to savings in productivity costs, total societal costs were lower with use of the strategy involving MR imaging by a mean of $153+/-488 (P=.54). CONCLUSION: MR imaging can be used without additional costs or disadvantageous effects on function to obviate arthroscopy in patients with nonacute knee symptoms who are highly suspected of having intraarticular knee abnormality.


Sujet(s)
Coûts des soins de santé/statistiques et données numériques , Maladies articulaires/diagnostic , Maladies articulaires/économie , Articulation du genou/anatomopathologie , Imagerie par résonance magnétique/économie , Imagerie par résonance magnétique/statistiques et données numériques , Triage/méthodes , Adolescent , Adulte , Analyse coût-bénéfice , Femelle , Humains , Maladies articulaires/épidémiologie , Mâle , Adulte d'âge moyen , Pays-Bas/épidémiologie , Prévalence , Reproductibilité des résultats , Appréciation des risques/méthodes , Facteurs de risque , Sensibilité et spécificité , Triage/statistiques et données numériques
18.
Neuromuscul Disord ; 16(1): 4-13, 2006 Jan.
Article de Anglais | MEDLINE | ID: mdl-16378727

RÉSUMÉ

The identification of an ever increasing number of gene defects in patients with neuromuscular disorders has disclosed both marked phenotype and genotype variability and considerable disease overlap. In order to offer an economic strategy to characterise the molecular defect in patients with unclassified neuromuscular disorders, we designed DNA marker sets for linkage analysis of 62 distinct neuromuscular disorders gene loci, including all known muscular dystrophies, congenital myopathies, congenital myasthenic syndromes and myotonias. Genotyping of marker loci of 140 clinically well-characterised families with unclassified neuromuscular disorders reduced the number of candidates to one or two genes in 49 % of the families. Subsequent mutation analysis and genome-wide scans enabled the determination of the genetic defect in 31 % of the families including the identification of a new gene and a new mutation in an unexpected candidate gene. This highlights the effective application of this approach both for diagnostic strategies as well as for the identification of new loci and genes.


Sujet(s)
Hétérogénéité génétique , Techniques de diagnostic moléculaire/méthodes , Protéines du muscle/génétique , Maladies neuromusculaires/diagnostic , Maladies neuromusculaires/génétique , Analyse de mutations d'ADN/méthodes , Bases de données génétiques/statistiques et données numériques , Diagnostic différentiel , Santé de la famille , Génotype , Humains , Techniques de diagnostic moléculaire/économie , Maladies neuromusculaires/classification
19.
Eur Radiol ; 16(1): 97-107, 2006 Jan.
Article de Anglais | MEDLINE | ID: mdl-15841383

RÉSUMÉ

The aim of this study is to evaluate the relation between bone bruise and (peri-)articular derangement and to assess the impact of bone bruise on presentation and short term course of knee complaints. We recorded MR abnormalities in 664 consecutive patients with sub-acute knee complaints. Patients were divided in four groups: patients with and without intra-articular knee pathology, subdivided in patients with and without bone bruise. We assessed function and symptoms at the time of MR and 6 months thereafter. Bone bruises were diagnosed in 124 of 664 patients (18.7%). Patients with bone bruise had significantly more complete ACL, lateral meniscal, MCL and LCL tears. Both with and without intra-articular pathology patients with bone bruise had a significantly poorer function at the time of MR (Noyes score of, respectively, 313.21 versus 344.81 and 306.98 versus 341.19). Patients with bone bruise and intra-articular pathology showed significantly more decrease in activity (decrease of Tegner score from 6.28 to 2.12 versus 5.70-2.55). At 6 months there were no significant differences in clinical parameters between the four groups. We concluded that bone bruise in combination with MCL tear is an important cause of initial clinical impairment in patients with sub-acute knee complaints. Clinical improvement within 6 months is more pronounced than in patients without bone bruise.


Sujet(s)
Maladies osseuses/diagnostic , Contusions/complications , Traumatismes du genou/diagnostic , Articulation du genou/anatomopathologie , Articulation du genou/chirurgie , Adolescent , Adulte , Arthroscopie/méthodes , Maladies osseuses/complications , Maladies du cartilage/complications , Maladies du cartilage/diagnostic , Femelle , Études de suivi , Humains , Traitement d'image par ordinateur/méthodes , Traumatismes du genou/complications , Imagerie par résonance magnétique/méthodes , Mâle , Adulte d'âge moyen , Études prospectives
20.
Acta Myol ; 25(2): 73-6, 2006 Oct.
Article de Anglais | MEDLINE | ID: mdl-18593008

RÉSUMÉ

We report on two unrelated patients clinically presenting with late-onset progressive limb girdle weakness; cardiomyopathy was seen in one patient. Muscle biopsy revealed a necrotic myopathy with numerous rimmed vacuoles, ultrastructurally typical paired-helical filaments, and reduced immunohistochemical staining for alpha-dystroglycan. Quadriceps sparing hereditary inclusion body myopathy due to mutations in GNE gene, and OPMD due to PABPN1 mutations were excluded, genetically. We detected a homozygous mutation of the FKRP gene (826C>A) in both patients. Mutations of FKRP have been reported in congenital muscular dystrophies, LGMD2I, cardiomyopathy and hyperCKemia, but not in myopathies with vacuoles and paired-helical filaments. Therefore, our findings further extend the morphological variability of muscular dystrophies due to FKRP mutations.


Sujet(s)
Dystrophies musculaires des ceintures/génétique , Dystrophies musculaires des ceintures/anatomopathologie , Protéines/génétique , Adulte , Cytosquelette/anatomopathologie , Humains , Mâle , Adulte d'âge moyen , Pentosyltransferases , Vacuoles/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE