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










Publication year range
2.
Med Sci (Paris) ; 39 Hors série n° 1: 72-73, 2023 11.
Article in French | MEDLINE | ID: mdl-37975777
3.
J Neuromuscul Dis ; 10(1): 125-133, 2023.
Article in English | MEDLINE | ID: mdl-36373293

ABSTRACT

We report three siblings from a non-consanguineous family presenting with contractural limb-girdle phenotype with intrafamilial variability. Muscle MRI showed posterior thigh and quadriceps involvement with a sandwich-like sign. Whole-exome sequencing identified two compound heterozygous missense TTN variants and one heterozygous LAMA2 variant. Brain MRI performed because of concentration difficulties in one of the siblings evidenced white-matter abnormalities, subsequently found in the others. The genetic analysis was re-oriented, revealing a novel pathogenic intronic LAMA2 variant which confirmed the LAMA2-RD diagnosis. This work highlights the importance of a thorough clinical phenotyping and the importance of brain imaging, in order to orientate and interpret the genetic analysis.


Subject(s)
Muscular Dystrophies, Limb-Girdle , Muscular Dystrophies , Humans , Muscular Dystrophies, Limb-Girdle/diagnostic imaging , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies/diagnostic imaging , Muscular Dystrophies/genetics , Brain/diagnostic imaging , Brain/pathology , Genetic Testing , Neuroimaging
4.
Front Genet ; 13: 1107969, 2022.
Article in English | MEDLINE | ID: mdl-36568380
6.
Clin Case Rep ; 9(9): e04128, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34484741

ABSTRACT

COL1-related overlap disorder is a condition, which is not yet considered as part of the 2017 EDS classification. However, it should be investigated as an alternative diagnosis for any patient with hypermobile EDS. This could allow providing appropriate genetic counseling.

7.
J Neuromuscul Dis ; 8(4): 633-645, 2021.
Article in English | MEDLINE | ID: mdl-33749658

ABSTRACT

BACKGROUND: Dominant and recessive autosomal pathogenic variants in the three major genes (COL6A1-A2-A3) encoding the extracellular matrix protein collagen VI underlie a group of myopathies ranging from early-onset severe conditions (Ullrich congenital muscular dystrophy) to milder forms maintaining independent ambulation (Bethlem myopathy). Diagnosis is based on the combination of clinical presentation, muscle MRI, muscle biopsy, analysis of collagen VI secretion, and COL6A1-A2-A3 genetic analysis, the interpretation of which can be challenging. OBJECTIVE: To refine the phenotypical spectrum associated with the frequent COL6A3 missense variant c.7447A>G (p.Lys2483Glu). METHODS: We report the clinical and molecular findings in 16 patients: 12 patients carrying this variant in compound heterozygosity with another COL6A3 variant, and four homozygous patients. RESULTS: Patients carrying this variant in compound heterozygosity with a truncating COL6A3 variant exhibit a phenotype consistent with COL6-related myopathies (COL6-RM), with joint contractures, proximal weakness and skin abnormalities. All remain ambulant in adulthood and only three have mild respiratory involvement. Most show typical muscle MRI findings. In five patients, reduced collagen VI secretion was observed in skin fibroblasts cultures. All tested parents were unaffected heterozygous carriers. Conversely, two out of four homozygous patients did not present with the classical COL6-RM clinical and imaging findings. Collagen VI immunolabelling on cultured fibroblasts revealed rather normal secretion in one and reduced secretion in another. Muscle biopsy from one homozygous patient showed myofibrillar disorganization and rimmed vacuoles. CONCLUSIONS: In light of our results, we postulate that the COL6A3 variant c.7447A>G may act as a modulator of the clinical phenotype. Thus, in patients with a typical COL6-RM phenotype, a second variant must be thoroughly searched for, while for patients with atypical phenotypes further investigations should be conducted to exclude alternative causes. This works expands the clinical and molecular spectrum of COLVI-related myopathies.


Subject(s)
Collagen Type VI/genetics , Muscular Dystrophies/genetics , Procollagen/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Heterozygote , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Muscle, Skeletal/pathology , Muscular Diseases/genetics , Mutation , Phenotype , Young Adult
8.
J Neuromuscul Dis ; 8(2): 273-285, 2021.
Article in English | MEDLINE | ID: mdl-33337382

ABSTRACT

A family of five male siblings (three survivors at 48, 53 and 58 years old; two deceased at 8 months old and 2.5 years old) demonstrating significant phenotypic variability ranging from intermediate to the myosclerotic like Bethlem myopathy is presented. Whole-exome sequencing (WES) identified a new homozygous missense mutation chr21:47402679 T > C in the canonical splice donor site of the second intron (c.227 + 2T>C) in the COL6A1 gene. mRNA analysis confirmed skipping of exon 2 encoding 925 amino-acids in 94-95% of resulting transcripts. Three sibs presented with intermediate phenotype of collagen VI-related dystrophies (48, 53 and 2.5 years old) while the fourth sibling (58 years old) was classified as Bethlem myopathy with spine rigidity. The two older siblings with the moderate progressive phenotype (48 and 53 years old) lost their ability to maintain a vertical posture caused by pronounced contractures of large joints, but continued to ambulate throughout life on fully bent legs without auxiliary means of support. Immunofluorescence analysis of dermal fibroblasts demonstrated that no type VI collagen was secreted in any of the siblings' cells, regardless of clinical manifestations severity while fibroblast proliferation and colony formation ability was decreased. The detailed genetic and long term clinical data contribute to broadening the genotypic and phenotypic spectrum of COL6A1 related disease.


Subject(s)
Collagen Type VI , Contracture/genetics , Muscular Dystrophies/congenital , Biological Variation, Population , Exons , Genotype , Humans , Infant , Introns , Male , Middle Aged , Muscular Dystrophies/genetics , Mutation , Mutation, Missense , Phenotype
9.
Antioxidants (Basel) ; 9(3)2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32197453

ABSTRACT

Congenital muscular dystrophy with laminin α2 chain-deficiency (LAMA2-CMD) is a severe neuromuscular disorder without a cure. Using transcriptome and proteome profiling as well as functional assays, we previously demonstrated significant metabolic impairment in skeletal muscle from LAMA2-CMD patients and mouse models. Reactive oxygen species (ROS) increase when oxygen homeostasis is not maintained and, here, we investigate whether oxidative stress indeed is involved in the pathogenesis of LAMA2-CMD. We also analyze the effects of two antioxidant molecules, N-acetyl-L-cysteine (NAC) and vitamin E, on disease progression in the dy2J/dy2J mouse model of LAMA2-CMD. We demonstrate increased ROS levels in LAMA2-CMD mouse and patient skeletal muscle. Furthermore, NAC treatment (150 mg/kg IP for 6 days/week for 3 weeks) led to muscle force loss prevention, reduced central nucleation and decreased the occurrence of apoptosis, inflammation, fibrosis and oxidative stress in LAMA2-CMD muscle. In addition, vitamin E (40 mg/kg oral gavage for 6 days/week for 2 weeks) improved morphological features and reduced inflammation and ROS levels in dy2J/dy2J skeletal muscle. We suggest that NAC and to some extent vitamin E might be potential future supportive treatments for LAMA2-CMD as they improve numerous pathological hallmarks of LAMA2-CMD.

13.
JCI Insight ; 4(6)2019 03 21.
Article in English | MEDLINE | ID: mdl-30895940

ABSTRACT

The clinical application of advanced next-generation sequencing technologies is increasingly uncovering novel classes of mutations that may serve as potential targets for precision medicine therapeutics. Here, we show that a deep intronic splice defect in the COL6A1 gene, originally discovered by applying muscle RNA sequencing in patients with clinical findings of collagen VI-related dystrophy (COL6-RD), inserts an in-frame pseudoexon into COL6A1 mRNA, encodes a mutant collagen α1(VI) protein that exerts a dominant-negative effect on collagen VI matrix assembly, and provides a unique opportunity for splice-correction approaches aimed at restoring normal gene expression. Using splice-modulating antisense oligomers, we efficiently skipped the pseudoexon in patient-derived fibroblast cultures and restored a wild-type matrix. Similarly, we used CRISPR/Cas9 to precisely delete an intronic sequence containing the pseudoexon and efficiently abolish its inclusion while preserving wild-type splicing. Considering that this splice defect is emerging as one of the single most frequent mutations in COL6-RD, the design of specific and effective splice-correction therapies offers a promising path for clinical translation.


Subject(s)
Collagen Type VI/genetics , Genetic Predisposition to Disease/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , RNA Splicing , Base Sequence , CRISPR-Cas Systems , DNA Mutational Analysis , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Genetic Therapy/methods , High-Throughput Nucleotide Sequencing , Humans , Introns/genetics , Mutation , RNA Splice Sites , RNA, Messenger/metabolism , Skin/pathology
16.
Am J Pathol ; 187(3): 654-664, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28082118

ABSTRACT

Fibrosis is the main complication of muscular dystrophies. We identified moesin, a member of the ezrin-radixin-moesin family, in dystrophic muscles of mice representing Duchenne and congenital muscular dystrophies (DMD and CMD, respectively) and dysferlinopathy, but not in the wild type. High levels of moesin were also observed in muscle biopsy specimens from DMD, Ullrich CMD, and merosin-deficient CMD patients, all of which present high levels of fibrosis. The myofibroblasts, responsible for extracellular matrix protein synthesis, and the macrophages infiltrating the dystrophic muscles were the source of moesin. Moesin-positive cells were embedded within the fibrotic areas between the myofibers adjacent to the collagen type I fibers. Radixin was also synthesized by the myofibroblasts, whereas ezrin colocalized with the myofiber membranes. In animal models and patients' muscles, part of the moesin was in its active phosphorylated form. Inhibition of fibrosis by halofuginone, an antifibrotic agent, resulted in a major decrease in moesin levels in the muscles of DMD and CMD mice. In summary, the results of this study may pave the way for exploiting moesin as a novel target for intervention in MDs, and as part of a battery of biomarkers to evaluate treatment success in preclinical studies and clinical trials.


Subject(s)
Muscular Dystrophies/metabolism , Adult , Animals , Child , Child, Preschool , Collagen Type I/metabolism , Cytoskeletal Proteins/metabolism , Diaphragm/drug effects , Diaphragm/metabolism , Homozygote , Humans , Immunohistochemistry , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Inbred mdx , Microfilament Proteins , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies/pathology , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphorylation/drug effects , Piperidines/pharmacology , Quinazolinones/pharmacology
17.
Am J Pathol ; 187(3): 505-516, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28056338

ABSTRACT

Collagen IV is a major component of basement membranes (BMs). The α1(IV) chain, encoded by the COL4A1 gene, is expressed ubiquitously and associates with the α2(IV) chain to form the α1α1α2(IV) heterotrimer. Several COL4A1 mutations affecting a conformational domain containing integrin-binding sites are responsible for the systemic syndrome of hereditary angiopathy, nephropathy, aneurysms, and cramps (HANAC). To analyze the pathophysiology of HANAC, Col4a1 mutant mice bearing the p.Gly498Val mutation were generated. Analysis of the skeletal muscles of Col4a1G498V mutant animals showed morphologic characteristics of a muscular dystrophy phenotype with myofiber atrophy, centronucleation, focal inflammatory infiltrates, and fibrosis. Abnormal ultrastructural aspects of muscle BMs was associated with reduced extracellular secretion of the mutant α1α1α2(IV) trimer. In addition to muscular dystrophic features, endothelial cell defects of the muscle capillaries were observed, with intracytoplasmic accumulation of the mutant α1α1α2(IV) molecules, endoplasmic reticulum cisternae dilation, and up-regulation of endoplasmic reticulum stress markers. Induction of the unfolded protein response in Col4a1 mutant muscle tissue resulted in an excess of apoptosis in endothelial cells. HANAC mutant animals also presented with a muscular functional impairment and increased serum creatine kinase levels reflecting altered muscle fiber sarcolemma. This extensive description of the muscular phenotype of the Col4a1 HANAC murine model suggests a potential contribution of primary endothelial cell defects, together with muscle BM alterations, to the development of COL4A1-related myopathy.


Subject(s)
Blood Vessels/abnormalities , Collagen Type IV/genetics , Muscle Cramp/genetics , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Mutation/genetics , Raynaud Disease/genetics , Animals , Apoptosis , Blood Vessels/pathology , Body Weight , Creatine Kinase/blood , Dystrophin/metabolism , Endoplasmic Reticulum Stress , Endothelial Cells/pathology , Endothelial Cells/ultrastructure , Extracellular Matrix/metabolism , Integrin beta1/metabolism , Mice , Mice, Mutant Strains , Muscle, Skeletal/ultrastructure , Organ Size , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
18.
J Neuromuscul Dis ; 3(2): 267-274, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27854213

ABSTRACT

The classical phenotypes of collagen VI-associated myopathies are well described. Little is known, however, about the progression of patients at the mildest end of the clinical spectrum. In this report, we describe the clinical findings and the results of MRI, muscle biopsy, collagen VI expression in cultured skin fibroblasts and genetic tests of a series of patients with Bethlem myopathy. Our series highlights the existence of mild presentations of this disorder that progresses only slightly and can easily be overlooked. Analysis of the genetic studies suggests that missense mutations can be associated to a milder clinical presentation. Muscle MRI is extremely useful as it shows a pathognomonic pattern in most patients, especially those with some degree of muscle weakness.


Subject(s)
Contracture/physiopathology , Muscular Dystrophies/congenital , Adult , Collagen Type VI/genetics , Collagen Type VI/metabolism , Contracture/diagnostic imaging , Contracture/genetics , Contracture/pathology , Disease Progression , Female , Fibroblasts/metabolism , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Middle Aged , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Muscular Dystrophies/diagnostic imaging , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Muscular Dystrophies/physiopathology , Mutation, Missense , Phenotype , Severity of Illness Index , Young Adult
19.
Hum Mol Genet ; 25(8): 1559-73, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-27008887

ABSTRACT

Despite recent progress in the genetic characterization of congenital muscle diseases, the genes responsible for a significant proportion of cases remain unknown. We analysed two branches of a large consanguineous family in which four patients presented with a severe new phenotype, clinically marked by neonatal-onset muscle weakness predominantly involving axial muscles, life-threatening respiratory failure, skin abnormalities and joint hyperlaxity without contractures. Muscle biopsies showed the unreported association of multi-minicores, caps and dystrophic lesions. Genome-wide linkage analysis followed by gene and exome sequencing in patients identified a homozygous nonsense mutation in TRIP4 encoding Activating Signal Cointegrator-1 (ASC-1), a poorly characterized transcription coactivator never associated with muscle or with human inherited disease. This mutation resulted in TRIP4 mRNA decay to around 10% of control levels and absence of detectable protein in patient cells. ASC-1 levels were higher in axial than in limb muscles in mouse, and increased during differentiation in C2C12 myogenic cells. Depletion of ASC-1 in cultured muscle cells from a patient and in Trip4 knocked-down C2C12 led to a significant reduction in myotube diameter ex vivo and in vitro, without changes in fusion index or markers of initial myogenic differentiation. This work reports the first TRIP4 mutation and defines a novel form of congenital muscle disease, expanding their histological, clinical and molecular spectrum. We establish the importance of ASC-1 in human skeletal muscle, identify transcriptional co-regulation as novel pathophysiological pathway, define ASC-1 as a regulator of late myogenic differentiation and suggest defects in myotube growth as a novel myopathic mechanism.


Subject(s)
Codon, Nonsense , Muscle Development , Muscular Diseases/congenital , Muscular Diseases/pathology , Transcription Factors/genetics , Adolescent , Animals , Cell Differentiation , Cell Line , Child , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Infant , Male , Mice , Muscle, Skeletal/metabolism , Muscular Diseases/genetics , Pedigree , RNA Stability , Sequence Analysis, DNA , Transcription Factors/metabolism
20.
Med Sci (Paris) ; 31 Spec No 3: 28-9, 2015 Nov.
Article in French | MEDLINE | ID: mdl-26546928
SELECTION OF CITATIONS
SEARCH DETAIL
...