Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.531
Filter
2.
Sci Rep ; 14(1): 11225, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755190

ABSTRACT

Muscular dystrophies (MDs) are inherited genetic diseases causing weakness and degeneration of muscles. The distribution of muscle weakness differs between MDs, involving distal muscles or proximal muscles. While the mutations in most of the MD-associated genes lead to either distal or proximal onset, there are also genes whose mutations can cause both types of onsets. We hypothesized that the genes associated with different MD onsets code proteins with distinct cellular functions. To investigate this, we collected the MD-associated genes and assigned them to three onset groups: genes mutated only in distal onset dystrophies, genes mutated only in proximal onset dystrophies, and genes mutated in both types of onsets. We then systematically evaluated the cellular functions of these gene sets with computational strategies based on functional enrichment analysis and biological network analysis. Our analyses demonstrate that genes mutated in either distal or proximal onset MDs code proteins linked with two distinct sets of cellular processes. Interestingly, these two sets of cellular processes are relevant for the genes that are associated with both onsets. Moreover, the genes associated with both onsets display high centrality and connectivity in the network of muscular dystrophy genes. Our findings support the hypothesis that the proteins associated with distal or proximal onsets have distinct functional characteristics, whereas the proteins associated with both onsets are multifunctional.


Subject(s)
Muscle Weakness , Muscular Dystrophies , Mutation , Humans , Muscular Dystrophies/genetics , Muscle Weakness/genetics , Gene Regulatory Networks , Computational Biology/methods , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscle, Skeletal/pathology
3.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38749543

ABSTRACT

Phosphatidylcholine (PC) is the major membrane phospholipid in most eukaryotic cells. Bi-allelic loss of function variants in CHKB, encoding the first step in the synthesis of PC, is the cause of a rostrocaudal muscular dystrophy in both humans and mice. Loss of sarcolemma integrity is a hallmark of muscular dystrophies; however, how this occurs in the absence of choline kinase function is not known. We determine that in Chkb -/- mice there is a failure of the α7ß1 integrin complex that is specific to affected muscle. We observed that in Chkb -/- hindlimb muscles there is a decrease in sarcolemma association/abundance of the PI(4,5)P2 binding integrin complex proteins vinculin, and α-actinin, and a decrease in actin association with the sarcolemma. In cells, pharmacological inhibition of choline kinase activity results in internalization of a fluorescent PI(4,5)P2 reporter from discrete plasma membrane clusters at the cell surface membrane to cytosol, this corresponds with a decreased vinculin localization at plasma membrane focal adhesions that was rescued by overexpression of CHKB.


Subject(s)
Choline Kinase , Integrins , Mice, Knockout , Muscular Dystrophies , Sarcolemma , Vinculin , Animals , Mice , Vinculin/metabolism , Vinculin/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Integrins/metabolism , Choline Kinase/metabolism , Choline Kinase/genetics , Sarcolemma/metabolism , Humans , Focal Adhesions/metabolism , Cell Membrane/metabolism , Actinin/metabolism , Actinin/genetics , Muscle, Skeletal/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Actins/metabolism , Disease Models, Animal
5.
Matrix Biol ; 129: 44-58, 2024 May.
Article in English | MEDLINE | ID: mdl-38582404

ABSTRACT

Extracellular matrix (ECM) pathologic remodeling underlies many disorders, including muscular dystrophy. Tissue decellularization removes cellular components while leaving behind ECM components. We generated "on-slide" decellularized tissue slices from genetically distinct dystrophic mouse models. The ECM of dystrophin- and sarcoglycan-deficient muscles had marked thrombospondin 4 deposition, while dysferlin-deficient muscle had excess decorin. Annexins A2 and A6 were present on all dystrophic decellularized ECMs, but annexin matrix deposition was excessive in dysferlin-deficient muscular dystrophy. Muscle-directed viral expression of annexin A6 resulted in annexin A6 in the ECM. C2C12 myoblasts seeded onto decellularized matrices displayed differential myoblast mobility and fusion. Dystrophin-deficient decellularized matrices inhibited myoblast mobility, while dysferlin-deficient decellularized matrices enhanced myoblast movement and differentiation. Myoblasts treated with recombinant annexin A6 increased mobility and fusion like that seen on dysferlin-deficient decellularized matrix and demonstrated upregulation of ECM and muscle cell differentiation genes. These findings demonstrate specific fibrotic signatures elicit effects on myoblast activity.


Subject(s)
Cell Differentiation , Cell Movement , Dysferlin , Extracellular Matrix , Myoblasts , Sarcoglycans , Animals , Myoblasts/metabolism , Myoblasts/cytology , Extracellular Matrix/metabolism , Mice , Sarcoglycans/genetics , Sarcoglycans/metabolism , Dysferlin/genetics , Dysferlin/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Dystrophin/genetics , Dystrophin/metabolism , Annexin A2/genetics , Annexin A2/metabolism , Decorin/genetics , Decorin/metabolism , Cell Line , Disease Models, Animal , Muscle, Skeletal/metabolism
6.
Physiol Rep ; 12(8): e16020, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38658362

ABSTRACT

Desminopathy R350P is a human myopathy that is characterized by the progressive loss of muscle fiber organization. This results in the loss of muscle size, mobility, and strength. In desminopathy, inflammation affects muscle homeostasis and repair, and contributes to progressive muscle deterioration. Mitochondria morphology was also suggested to affect desminopathy progression. Epicatechin (Epi)-a natural compound found in cacao-has been proposed to regulate inflammatory signaling and mitochondria morphology in human and animal models. Hence, we hypothesize chronic Epi consumption to improve inflammatory pathway and mitochondria morphology in the peripheral blood mononuclear cells (PBMCs) of a desminopathy R350P patient. We found that 12 weeks of Epi consumption partially restored TRL4 signaling, indicative of inflammatory signaling and mitochondria morphology in the desminopathy patient. Moreover, Epi consumption improved blood health parameters, including reduced HOMA-IR and IL-6 levels in the desminopathy patient. This indicates that Epi consumption could be a useful tool to slow disease progression in desminopathy patients.


Subject(s)
Catechin , Leukocytes, Mononuclear , Mitochondria , Humans , Catechin/pharmacology , Catechin/administration & dosage , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Male , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/drug therapy , Muscular Dystrophies/genetics , Adult , Female , Inflammation/metabolism , Inflammation/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/drug therapy , Desmin/metabolism , Desmin/genetics
7.
Neuromuscul Disord ; 38: 26-41, 2024 May.
Article in English | MEDLINE | ID: mdl-38554696

ABSTRACT

LMNA-related congenital muscular dystrophy (L-CMD) is caused by mutations in the LMNA gene, encoding lamin A/C. To further understand the molecular mechanisms of L-CMD, proteomic profiling using DIA mass spectrometry was conducted on immortalized myoblasts and myotubes from controls and L-CMD donors each harbouring a different LMNA mutation (R249W, del.32 K and L380S). Compared to controls, 124 and 228 differentially abundant proteins were detected in L-CMD myoblasts and myotubes, respectively, and were associated with enriched canonical pathways including synaptogenesis and necroptosis in myoblasts, and Huntington's disease and insulin secretion in myotubes. Abnormal nuclear morphology and reduced lamin A/C and emerin abundance was evident in all L-CMD cell lines compared to controls, while nucleoplasmic aggregation of lamin A/C was restricted to del.32 K cells, and mislocalization of emerin was restricted to R249W cells. Abnormal nuclear morphology indicates loss of nuclear lamina integrity as a common feature of L-CMD, likely rendering muscle cells vulnerable to mechanically induced stress, while differences between L-CMD cell lines in emerin and lamin A localization suggests that some molecular alterations in L-CMD are mutation specific. Nonetheless, identifying common proteomic alterations and molecular pathways across all three L-CMD lines has highlighted potential targets for the development of non-mutation specific therapies.


Subject(s)
Lamin Type A , Muscular Dystrophies , Proteomics , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Mutation , Myoblasts/metabolism , Male , Cell Line , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
8.
Biomolecules ; 14(3)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38540676

ABSTRACT

Dysferlinopathies refer to a spectrum of muscular dystrophies that cause progressive muscle weakness and degeneration. They are caused by mutations in the DYSF gene, which encodes the dysferlin protein that is crucial for repairing muscle membranes. This review delves into the clinical spectra of dysferlinopathies, their molecular mechanisms, and the spectrum of emerging therapeutic strategies. We examine the phenotypic heterogeneity of dysferlinopathies, highlighting the incomplete understanding of genotype-phenotype correlations and discussing the implications of various DYSF mutations. In addition, we explore the potential of symptomatic, pharmacological, molecular, and genetic therapies in mitigating the disease's progression. We also consider the roles of diet and metabolism in managing dysferlinopathies, as well as the impact of clinical trials on treatment paradigms. Furthermore, we examine the utility of animal models in elucidating disease mechanisms. By culminating the complexities inherent in dysferlinopathies, this write up emphasizes the need for multidisciplinary approaches, precision medicine, and extensive collaboration in research and clinical trial design to advance our understanding and treatment of these challenging disorders.


Subject(s)
Muscular Dystrophies, Limb-Girdle , Muscular Dystrophies , Animals , Muscle Proteins/genetics , Membrane Proteins/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/therapy , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies/genetics , Mutation
9.
Neurology ; 102(7): e209258, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38484275

ABSTRACT

We describe the case of a 19-month-old girl presenting with gross motor delays, hypotonia, diminished deep tendon reflexes, hyperCKaemia, extensive white matter changes on MRI brain, and electromyography studies consistent with myopathy. The differential diagnosis for infantile-onset hypotonia and muscle weakness is broad. It includes numerous subtypes of genetic disorders, including congenital muscular dystrophies, congenital myopathies, congenital myasthenic syndromes, spinal muscular atrophy, single-gene genetic syndromes, and inborn errors of metabolism. We outline our clinical approach leading to the diagnosis of a distinctive genetic neuromuscular condition essential for neurologists and geneticists working with patients of all ages to recognize.


Subject(s)
Muscular Diseases , Muscular Dystrophies , White Matter , Female , Humans , Infant , Muscle Hypotonia/etiology , White Matter/diagnostic imaging , Muscular Diseases/genetics , Muscular Dystrophies/genetics , Clinical Reasoning
10.
Nat Commun ; 15(1): 1758, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38413582

ABSTRACT

SNURPORTIN-1, encoded by SNUPN, plays a central role in the nuclear import of spliceosomal small nuclear ribonucleoproteins. However, its physiological function remains unexplored. In this study, we investigate 18 children from 15 unrelated families who present with atypical muscular dystrophy and neurological defects. Nine hypomorphic SNUPN biallelic variants, predominantly clustered in the last coding exon, are ascertained to segregate with the disease. We demonstrate that mutant SPN1 failed to oligomerize leading to cytoplasmic aggregation in patients' primary fibroblasts and CRISPR/Cas9-mediated mutant cell lines. Additionally, mutant nuclei exhibit defective spliceosomal maturation and breakdown of Cajal bodies. Transcriptome analyses reveal splicing and mRNA expression dysregulation, particularly in sarcolemmal components, causing disruption of cytoskeletal organization in mutant cells and patient muscle tissues. Our findings establish SNUPN deficiency as the genetic etiology of a previously unrecognized subtype of muscular dystrophy and provide robust evidence of the role of SPN1 for muscle homeostasis.


Subject(s)
Muscular Dystrophies , Child , Humans , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , RNA/metabolism , RNA Splicing/genetics , Spliceosomes/genetics , Spliceosomes/metabolism
11.
Life Sci ; 341: 122489, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38340979

ABSTRACT

Lamins are inner nuclear membrane proteins that belong to the intermediate filament family. Lamin A/C lie adjacent to the heterochromatin structure in polymer form, providing skeletal to the nucleus. Based on the localization, lamin A/C provides nuclear stability and cytoskeleton to the nucleus and modulates chromatin organization and gene expression. Besides being the structural protein making the inner nuclear membrane in polymer form, lamin A/C functions as a signalling molecule involved in gene expression as an enhancer inside the nucleus. Lamin A/C regulates various cellular pathways like autophagy and energy balance in the cytoplasm. Its expression is highly variable in differentiated tissues, higher in hard tissues like bone and muscle cells, and lower in soft tissues like the liver and brain. In muscle cells, including the heart, lamin A/C must be expressed in a balanced state. Lamin A/C mutation is linked with various diseases, such as muscular dystrophy, lipodystrophy, and cardiomyopathies. It has been observed that a good number of mutations in the LMNA gene impact cardiac activity and its function. Although several works have been published, there are still several unexplored areas left regarding the lamin A/C function and structure in the cardiovascular system and its pathological state. In this review, we focus on the structural organization, expression pattern, and function of lamin A/C, its interacting partners, and the pathophysiology associated with mutations in the lamin A/C gene, with special emphasis on cardiovascular diseases. With the recent finding on lamin A/C, we have summarized the possible therapeutic interventions to treat cardiovascular symptoms and reverse the molecular changes.


Subject(s)
Cardiomyopathies , Muscular Dystrophies , Humans , Lamin Type A/genetics , Lamin Type A/chemistry , Lamin Type A/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Polymers
12.
Cell Rep ; 43(2): 113720, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38308845

ABSTRACT

LMNA gene mutation can cause muscular dystrophy, and post-translational modification plays a critical role in regulating its function. Here, we identify that lamin A is palmitoylated at cysteine 522, 588, and 591 residues, which are reversely catalyzed by palmitoyltransferase zinc finger DHHC-type palmitoyltransferase 5 (ZDHHC5) and depalmitoylase α/ß hydrolase domain 7 (ABHD7). Furthermore, the metabolite lactate promotes palmitoylation of lamin A by inhibiting the interaction between it and ABHD7. Interestingly, low-level palmitoylation of lamin A promotes, whereas high-level palmitoylation of lamin A inhibits, murine myoblast differentiation. Together, these observations suggest that ABHD7-mediated depalmitoylation of lamin A controls myoblast differentiation.


Subject(s)
Lamin Type A , Muscular Dystrophies , Animals , Mice , Cell Differentiation , Lamin Type A/metabolism , Muscular Dystrophies/genetics , Myoblasts/metabolism , Protein Processing, Post-Translational
13.
Neuromuscul Disord ; 36: 16-22, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38306718

ABSTRACT

The European Joint Programme on Rare Diseases (EJPRD) funded the workshop "LAMA2-Muscular Dystrophy: Paving the road to therapy", bringing together 40 health-care professionals, researchers, patient-advocacy groups, Early-Career Scientists and other stakeholders from 14 countries. Progress in natural history, pathophysiology, trial readiness, and treatment strategies was discussed together with efforts to increase patient-awareness and strengthen collaborations. Key outcomes were (a) ongoing natural history studies in 7 countries already covered more than 350 patients. The next steps are to include additional countries, harmonise data collection and define a minimal dataset; (b) therapy development was largely complementary. Approaches included LAMA2-replacement and correction, LAMA1-reactivation, mRNA modulation, linker-protein expression, targeting downstream processes and identifying modifiers, using viral vectors, muscle stem cells, iPSC and mouse models and patient lines; (c) LAMA2-Europe will inform patients (-representatives) worldwide on standards of care and scientific progress, and enable sharing experiences. Follow-up monthly online meetings and research repositories have been established to create sustainable collaborations.


Subject(s)
Muscular Dystrophies , Rare Diseases , Mice , Animals , Humans , Spain , Rare Diseases/genetics , Rare Diseases/therapy , Laminin/genetics , Laminin/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Europe
14.
Neurogenetics ; 25(2): 93-102, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38296890

ABSTRACT

Congenital muscular dystrophies (CMDs) are a group of rare muscle disorders characterized by early onset hypotonia and motor developmental delay associated with brain malformations with or without eye anomalies in the most severe cases. In this study, we aimed to uncover the genetic basis of severe CMD in Egypt and to determine the efficacy of whole exome sequencing (WES)-based genetic diagnosis in this population. We recruited twelve individuals from eleven families with a clinical diagnosis of CMD with brain malformations that fell into two groups: seven patients with suspected dystroglycanopathy and five patients with suspected merosin-deficient CMD. WES was analyzed by variant filtering using multiple approaches including splicing and copy number variant (CNV) analysis. We identified likely pathogenic variants in FKRP in two cases and variants in POMT1, POMK, and B3GALNT2 in three individuals. All individuals with merosin-deficient CMD had truncating variants in LAMA2. Further analysis in one of the two unsolved cases showed a homozygous protein-truncating variant in Feline Leukemia Virus subgroup C Receptor 1 (FLVCR1). FLVCR1 loss of function has never been previously reported. Yet, loss of function of its paralog, FLVCR2, causes lethal hydranencephaly-hydrocephaly syndrome (Fowler Syndrome) which should be considered in the differential diagnosis for dystroglycanopathy. Overall, we reached a diagnostic rate of 86% (6/7) for dystroglycanopathies and 100% (5/5) for merosinopathy. In conclusion, our results provide further evidence that WES is an important diagnostic method in CMD in developing countries to improve the diagnostic rate, management plan, and genetic counseling for these disorders.


Subject(s)
Brain , Exome Sequencing , Muscular Dystrophies , N-Acetylglucosaminyltransferases , Humans , Male , Egypt , Female , Muscular Dystrophies/genetics , Muscular Dystrophies/diagnosis , Child, Preschool , Brain/abnormalities , Brain/pathology , Child , Infant , Laminin/genetics , Receptors, Virus/genetics , Mannosyltransferases/genetics , Pedigree , Pentosyltransferases/genetics , DNA Copy Number Variations , Mutation , Adolescent , Nervous System Malformations/genetics
15.
Eur J Hum Genet ; 32(3): 342-349, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38177406

ABSTRACT

DAG1 encodes for dystroglycan, a key component of the dystrophin-glycoprotein complex (DGC) with a pivotal role in skeletal muscle function and maintenance. Biallelic loss-of-function DAG1 variants cause severe muscular dystrophy and muscle-eye-brain disease. A possible contribution of DAG1 deficiency to milder muscular phenotypes has been suggested. We investigated the genetic background of twelve subjects with persistent mild-to-severe hyperCKemia to dissect the role of DAG1 in this condition. Genetic testing was performed through exome sequencing (ES) or custom NGS panels including various genes involved in a spectrum of muscular disorders. Histopathological and Western blot analyses were performed on muscle biopsy samples obtained from three patients. We identified seven novel heterozygous truncating variants in DAG1 segregating with isolated or pauci-symptomatic hyperCKemia in all families. The variants were rare and predicted to lead to nonsense-mediated mRNA decay or the formation of a truncated transcript. In four cases, DAG1 variants were inherited from similarly affected parents. Histopathological analysis revealed a decreased expression of dystroglycan subunits and Western blot confirmed a significantly reduced expression of beta-dystroglycan in muscle samples. This study supports the pathogenic role of DAG1 haploinsufficiency in isolated or pauci-symptomatic hyperCKemia, with implications for clinical management and genetic counseling.


Subject(s)
Muscular Diseases , Muscular Dystrophies , Humans , Dystroglycans/genetics , Dystroglycans/metabolism , Haploinsufficiency , Muscular Dystrophies/genetics , Muscle, Skeletal/pathology , Muscular Diseases/pathology
16.
JCI Insight ; 9(3)2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38175727

ABSTRACT

The Murphy Roths Large (MRL) mouse strain has "super-healing" properties that enhance recovery from injury. In mice, the DBA/2J strain intensifies many aspects of muscular dystrophy, so we evaluated the ability of the MRL strain to suppress muscular dystrophy in the Sgcg-null mouse model of limb girdle muscular dystrophy. A comparative analysis of Sgcg-null mice in the DBA/2J versus MRL strains showed greater myofiber regeneration, with reduced structural degradation of muscle in the MRL strain. Transcriptomic profiling of dystrophic muscle indicated strain-dependent expression of extracellular matrix (ECM) and TGF-ß signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized myoscaffolds. Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-ß1 and TGF-ß3 throughout the matrix. Dystrophic myoscaffolds from the MRL background, but not the DBA/2J background, were enriched in myokines like IGF-1 and IL-6. C2C12 myoblasts seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J muscles showed the MRL background induced greater myoblast differentiation compared with dystrophic DBA/2J myoscaffolds. Thus, the MRL background imparts its effect through a highly regenerative ECM, which is active even in muscular dystrophy.


Subject(s)
Muscular Dystrophies, Limb-Girdle , Muscular Dystrophies , Mice , Animals , Mice, Inbred DBA , Muscular Dystrophies/genetics , Muscles , Extracellular Matrix , Mice, Knockout
18.
J Invest Dermatol ; 144(3): 547-562.e9, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37716646

ABSTRACT

Plectin, a highly versatile and multifunctional cytolinker, has been implicated in several multisystemic disorders. Most sequence variations in the human plectin gene (PLEC) cause epidermolysis bullosa simplex with muscular dystrophy (EBS-MD), an autosomal recessive skin-blistering disorder associated with progressive muscle weakness. In this study, we performed a comprehensive cell biological analysis of dermal fibroblasts from three different patients with EBS-MD, where PLEC expression analyses revealed preserved mRNA levels in all cases, whereas full-length plectin protein content was significantly reduced or completely absent. Downstream effects of pathogenic PLEC sequence alterations included massive bundling of vimentin intermediate filament networks, including the occurrence of ring-like nuclei-encasing filament bundles, elongated mitochondrial networks, and abnormal nuclear morphologies. We found that essential fibroblast functions such as wound healing, migration, or orientation upon cyclic stretch were significantly impaired in the cells of patients with EBS-MD. Finally, EBS-MD fibroblasts displayed reduced adhesion capacities, which could be attributed to smaller focal adhesion contacts. Our study not only emphasizes plectin's functional role in human skin fibroblasts, it also provides further insights into the understanding of EBS-MD-associated disease mechanisms.


Subject(s)
Epidermolysis Bullosa Simplex , Muscular Dystrophies, Limb-Girdle , Muscular Dystrophies , Humans , Intermediate Filaments/metabolism , Plectin/genetics , Epidermolysis Bullosa Simplex/pathology , Muscular Dystrophies/complications , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Mitochondria/metabolism , Fibroblasts/metabolism , Intermediate Filament Proteins/metabolism
20.
Am J Med Genet A ; 194(3): e63462, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37929330

ABSTRACT

We describe a family with two maternal half-brothers both of whom presented with muscular dystrophy, autism spectrum disorder, developmental delay, and sensorineural hearing loss. The elder brother had onset of features at ~3 months of age, followed by clinical confirmation of muscular dystrophy at 3 years. Skeletal biopsy staining at 4.7 years showed an absence of dystrophin protein which prompted extensive molecular testing over 4 years that included gene panels, targeted single-gene assays, arrays, and karyotyping, all of which failed to identify a clinically significant variant in the DMD gene. At 10 years of age, clinical whole-genome sequencing (cWGS) was performed, which revealed a novel hemizygous ~50.7 Mb balanced pericentric inversion on chromosome X that disrupts the DMD gene in both siblings, consistent with the muscular dystrophy phenotype. This inversion also impacts the upstream regulatory region of POU3F4, structural rearrangements which are known to cause hearing loss. The unaffected mother is a heterozygous carrier for the pericentric inversion. This finding illustrates the ability of cWGS to detect a wide breadth of disease-causing genomic variations including large genomic rearrangements.


Subject(s)
Autism Spectrum Disorder , Muscular Dystrophies , Muscular Dystrophy, Duchenne , Child, Preschool , Female , Humans , Male , Autism Spectrum Disorder/genetics , Base Sequence , Chromosome Inversion/genetics , Dystrophin/genetics , Muscular Dystrophies/genetics , Muscular Dystrophy, Duchenne/genetics , POU Domain Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...