Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.276
Filter
1.
Nat Commun ; 15(1): 7965, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261465

ABSTRACT

Current adeno-associated virus (AAV) gene therapy using nature-derived AAVs is limited by non-optimal tissue targeting. In the treatment of muscular diseases (MD), high doses are often required but can lead to severe adverse effects. Here, we rationally design an AAV capsid that specifically targets skeletal muscle to lower treatment doses. We computationally integrate binding motifs of human integrin alphaV beta6, a skeletal muscle receptor, into a liver-detargeting capsid. Designed AAVs show higher productivity and superior muscle transduction compared to their parent. One variant, LICA1, demonstrates comparable muscle transduction to other myotropic AAVs with reduced liver targeting. LICA1's myotropic properties are observed across species, including non-human primate. Consequently, LICA1, but not AAV9, effectively delivers therapeutic transgenes and improved muscle functionality in two mouse MD models (male mice) at a low dose (5E12 vg/kg). These results underline the potential of our design method for AAV engineering and LICA1 variant for MD gene therapy.


Subject(s)
Dependovirus , Genetic Therapy , Muscle, Skeletal , Dependovirus/genetics , Animals , Humans , Muscle, Skeletal/metabolism , Mice , Genetic Therapy/methods , Male , Genetic Vectors/genetics , Integrins/metabolism , Integrins/genetics , Capsid Proteins/genetics , Capsid Proteins/metabolism , Muscular Diseases/therapy , Muscular Diseases/genetics , Transduction, Genetic , Liver/metabolism , Capsid/metabolism , Receptors, Vitronectin/metabolism , Receptors, Vitronectin/genetics , Disease Models, Animal , HEK293 Cells , Transgenes , Mice, Inbred C57BL , Antigens, Neoplasm
2.
Cells ; 13(17)2024 Sep 08.
Article in English | MEDLINE | ID: mdl-39273074

ABSTRACT

CCDC78 was identified as a novel candidate gene for autosomal dominant centronuclear myopathy-4 (CNM4) approximately ten years ago. However, to date, only one family has been described, and the function of CCDC78 remains unclear. Here, we analyze for the first time a family harboring a CCDC78 nonsense mutation to better understand the role of CCDC78 in muscle. METHODS: We conducted a comprehensive histopathological analysis on muscle biopsies, including immunofluorescent assays to detect multiple sarcoplasmic proteins. We examined CCDC78 transcripts and protein using WB in CCDC78-mutated muscle tissue; these analyses were also performed on muscle, lymphocytes, and fibroblasts from healthy subjects. Subsequently, we conducted RT-qPCR and transcriptome profiling through RNA-seq to evaluate changes in gene expression associated with CCDC78 dysfunction in muscle. Lastly, coimmunoprecipitation (Co-Ip) assays and mass spectrometry (LC-MS/MS) studies were carried out on extracted muscle proteins from both healthy and mutated subjects. RESULTS: The histopathological features in muscle showed novel histological hallmarks, which included areas of dilated and swollen sarcoplasmic reticulum (SR). We provided evidence of nonsense-mediated mRNA decay (NMD), identified the presence of novel CCDC78 transcripts in muscle and lymphocytes, and identified 1035 muscular differentially expressed genes, including several involved in the SR. Through the Co-Ip assays and LC-MS/MS studies, we demonstrated that CCDC78 interacts with two key SR proteins: SERCA1 and CASQ1. We also observed interactions with MYH1, ACTN2, and ACTA1. CONCLUSIONS: Our findings provide insight, for the first time, into the interactors and possible role of CCDC78 in skeletal muscle, locating the protein in the SR. Furthermore, our data expand on the phenotype previously associated with CCDC78 mutations, indicating potential histopathological hallmarks of the disease in human muscle. Based on our data, we can consider CCDC78 as the causative gene for CNM4.


Subject(s)
Muscle Proteins , Muscular Diseases , Humans , Male , Female , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Adult , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Pedigree , Middle Aged , Sarcoplasmic Reticulum/metabolism , Mutation/genetics , Nonsense Mediated mRNA Decay/genetics
3.
BMC Neurol ; 24(1): 312, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232665

ABSTRACT

BACKGROUND: Diagnosis of hereditary myopathy is often challenging owing to overlapping clinical phenotypes and muscle histopathological findings. This retrospective study aimed to identify the phenotypic and genotypic spectra of hereditary myopathies at a tertiary hospital in Riyadh, Saudi Arabia. METHODS: We reviewed the medical records of patients with hereditary myopathy who were evaluated between January 2018 and December 2022. RESULTS: Eighty-seven patients (78 families) were included, two-thirds were men with a mean age of 35 (SD 14.2) years. Limb-girdle muscular dystrophy (LGMD) was the most prevalent clinical diagnosis (25 cases; 29%), of whom, a genetic diagnosis was achieved in 15 of 22 patients tested (68%). In genetically confirmed LGMD, the most prevalent disorders were dysferlinopathy (27%) followed by fukutin-related protein (FKRP) - related limb girdle muscular dystrophy (20%), sarcoglycanopathy (20%), lamin A/C related myopathy (13%), and calpain-3 myopathy (13%). In 26 patients with pathogenic/likely pathogenic variants, the genetic testing method was whole exome sequencing (WES) (42%), Next generation sequencing (NGS) (31%), and targeted single gene analysis (27%). The sensitivity of each genetic testing method was as follows: 100% for targeted single-gene analysis, 100% for targeted analysis of D4Z4 repeat array units, 88% for myotonic dystrophy protein kinase (DMPK) repeat expansion analysis, 42% for NGS-neuromuscular panel, and 46% for WES. CONCLUSION: The prevalent types of hereditary myopathies were consistent with those reported locally and internationally. This study highlights the diagnostic yield of various molecular genetic tests for the diagnosis of hereditary myopathy in an adult cohort and the need for improved access to advanced molecular testing in cases suspected to have facioscapulohumeral muscular dystrophy (FSHD) or mitochondrial myopathies.


Subject(s)
Genetic Testing , Muscular Dystrophies, Limb-Girdle , Humans , Male , Saudi Arabia/epidemiology , Adult , Female , Middle Aged , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/epidemiology , Retrospective Studies , Genetic Testing/methods , Young Adult , Cohort Studies , Adolescent , Muscular Diseases/genetics , Muscular Diseases/diagnosis , Exome Sequencing/methods
4.
Mol Genet Genomic Med ; 12(9): e70003, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39248612

ABSTRACT

BACKGROUND: Newborn screening (NBS) for primary carnitine deficiency (PCD) has poor performance. This study aimed to evaluate the feasibility of incorporating next-generation sequencing (NGS) as a second-tier PCD test. METHODS: Between March and December 2020, 60,070 newborns were screened for inherited metabolic disorders. Newborns with free carnitine (C0) levels below 8.5 µmol/L were selected for second-tier genetic testing. RESULTS: In total, 130 (0.22%) newborns with low C0 levels underwent second-tier genetic testing, 87 (66.92%) had positive genetic testing results, and 30 (23.08%) carried pathogenic variants of the SLC22A5 gene. Six newborns were diagnosed with PCD. The incidence of PCD was approximately 1 in 1:10,012 newborns. The PPV reached 20% after combining with second-tier NGS. Of the eight variants identified in patients with PCD, the three most common variants were c.760C>T (p.Arg254*), c.51C>G (p.Phe17Leu), and c.1400C>G (p.Ser467Cys). The C0 levels of patients with PCD were significantly lower than those of PCD carriers (p = 0.0026) and PCD-negative individuals (p = 0.0005). CONCLUSIONS: Our results showed that the PPV reached 20% after combining with second-tier NGS. The MS/MS-based NBS and second-tier NGS combination can effectively reduce the false-positive rate and detect PCD in patients.


Subject(s)
Carnitine , Genetic Testing , High-Throughput Nucleotide Sequencing , Hyperammonemia , Solute Carrier Family 22 Member 5 , Humans , Carnitine/blood , Carnitine/deficiency , Solute Carrier Family 22 Member 5/genetics , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Hyperammonemia/genetics , Hyperammonemia/diagnosis , Infant, Newborn , Male , Female , Genetic Testing/methods , Genetic Testing/standards , Cardiomyopathies/genetics , Cardiomyopathies/diagnosis , Neonatal Screening/methods , Neonatal Screening/standards , Muscular Diseases/genetics , Muscular Diseases/diagnosis , Mutation
5.
Mitochondrion ; 78: 101945, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39134108

ABSTRACT

Mitochondria form a dynamic network within skeletal muscle. This network is not only responsible for producing adenosine triphosphate (ATP) through oxidative phosphorylation, but also responds through fission, fusion and mitophagy to various factors, such as increased energy demands, oxidative stress, inflammation, and calcium dysregulation. Mitochondrial dysfunction in skeletal muscle not only occurs in primary mitochondrial myopathies, but also other hereditary and acquired myopathies. As such, this review attempts to highlight the clinical and histopathologic aspects of mitochondrial dysfunction seen in hereditary and acquired myopathies, as well as discuss potential mechanisms leading to mitochondrial dysfunction and therapies to restore mitochondrial function.


Subject(s)
Muscular Diseases , Humans , Muscular Diseases/genetics , Muscular Diseases/pathology , Muscular Diseases/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria, Muscle/metabolism , Mitochondria, Muscle/pathology , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/pathology , Mitochondrial Myopathies/metabolism
6.
J Neuromuscul Dis ; 11(5): 1131-1137, 2024.
Article in English | MEDLINE | ID: mdl-39121134

ABSTRACT

HNRNPA1 variants are known to cause degenerative motoneuron and muscle diseases which manifests in middle age or later. We report on a girl with early childhood onset, rapidly progressive generalized myopathy including ultrastructural findings in line with a proteinopathy. Proteomics of patient-derived muscle and combined screening of genomic data for copy number variations identified a HNRNPA1 de novo intragenic deletion as causative for the phenotype. Our report expands the spectrum of HNRNPA1-related diseases towards early-childhood onset and adds HNRNPA1 to the growing list of ALS and myopathy genes for which certain mutations may cause severe pediatric phenotypes.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein A1 , Muscular Diseases , Humans , Female , Heterogeneous Nuclear Ribonucleoprotein A1/genetics , Muscular Diseases/genetics , Disease Progression , Age of Onset , Muscle, Skeletal/pathology , Phenotype , Mutation , Child
7.
J Clin Neuromuscul Dis ; 26(1): 16-31, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39163158

ABSTRACT

ABSTRACT: This update begins with a section on inflammatory myopathies covering inclusion body myositis in younger patients, the possibility of a pathogenic role for anti-cN1A antibodies, and a negative trial of arimoclomol in inclusion body myositis. The potential study of Janus kinase inhibitors in dermatomyositis is discussed as well as the possible role of targeted therapy for immune checkpoint inhibitor neuromuscular complications. Next, studies of disease-modifying or potential disease-modifying therapies for inherited myopathies are addressed including the encouraging follow-up study of gene replacement therapy for Duchenne muscular dystrophy (DMD), a negative trial of tamoxifen in DMD, and the complex topic of gene therapy for X-linked myotubular myopathy. A newly identified condition of muscular dystrophy from 3-hydroxy-3-methylglutaryl-CoA reductase mutations is addressed along with possible therapy. Other papers regarding GNE myopathy and long-term outcome of enzyme replacement therapy in infantile onset Pompe disease round out that section. Updates on the expanding spectra of anoctamin-5 myopathies, caveolinopathies, and congenital and mylagic myopathies from CACNA1S mutations follow as well as extensive discussion of Valosin containing protein proteinopathies, comprehensive management of Becker muscular dystrophy, and gastrointestinal complications in adult DMD.


Subject(s)
Muscular Diseases , Humans , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/therapy
8.
J Clin Neuromuscul Dis ; 26(1): 12-15, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39163157

ABSTRACT

ABSTRACT: X-linked myopathy with excessive autophagy is a rare disorder caused by a mutation in the vacuolar ATPase assembly factor gene which causes slowly progressive early onset proximal weakness and loss of ambulation by the age of 50-70 years. Electrodiagnostic (EDx) testing usually shows widespread complex repetitive and myotonic discharges, even in muscles unaffected clinically. We report a 65-year-old man who presented with progressive proximal weakness since his teenage years. Extensive workup over 30 years revealed inconclusive EDx and muscle histopathology findings. The diagnosis was finally made with genetic testing. Subsequent neuromuscular ultrasound was more informative of disease severity than repeat EDx and directed a muscle biopsy that showed an autophagic vacuolar myopathy and the novel identification of vacuoles in capillary endothelial cells. Although genetic testing is required for confirmation, in milder cases of X-linked myopathy with excessive autophagy, neuromuscular ultrasound may aid in diagnosis even when EDx findings are inconclusive.


Subject(s)
Genetic Diseases, X-Linked , Muscle, Skeletal , Muscular Diseases , Ultrasonography , Humans , Male , Aged , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/diagnostic imaging , Genetic Diseases, X-Linked/diagnosis , Muscular Diseases/genetics , Muscular Diseases/diagnostic imaging , Muscular Diseases/diagnosis , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Electrodiagnosis , Autophagy/genetics , Genetic Testing
9.
BMC Musculoskelet Disord ; 25(1): 661, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39174932

ABSTRACT

BACKGROUND: Neutral Lipid Storage Disease with Myopathy (NLSDM) is a rare lipid metabolism disorder caused by PNPLA2 gene mutations. Clinical manifestations are heterogeneous, and diagnosis is often delayed, usually gaining patients' attention due to the increased risk of cardiomyopathy. CASE PRESENTATION: We herein report a 36-year-old Asian male presenting with progressive limb weakness, muscle atrophy of limbs and trunk, dysarthria, and heart failure. Electromyography indicated myogenic changes, and muscle biopsy results revealed characteristics of lipid storage myopathy. Genetic analysis of PNPLA2 revealed two heterozygous mutations: c.757 + 1G > T (chr11-823588, splice-5) on intron 6 and c.919delG (chr11-823854, p.A307Pfs*13) on exon 7. The patient improved limb strength, and dysarthria disappeared after the Medium Chain Fatty Acids diet. CONCLUSIONS: In conclusion, we report for the first time that the two heterozygous mutations PNPLA2 c.919delG and c.757 + 1G > T together induced NLSDM, which was confirmed by muscle biopsy.


Subject(s)
Heterozygote , Lipase , Lipid Metabolism, Inborn Errors , Muscular Diseases , Mutation , Humans , Male , Lipase/genetics , Adult , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/diagnosis , Muscular Diseases/genetics , Muscular Diseases/diagnosis , Muscle, Skeletal/pathology , Acyltransferases
10.
Poult Sci ; 103(10): 104056, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39094498

ABSTRACT

Wooden breast (WB) is a myopathy mainly affecting pectoralis major (PM) muscle in modern commercial broiler chickens, causing enormous economic losses in the poultry industry. Recent studies have observed hepatic and PM muscle injury in broilers affected by WB, but the relationships between WB and the 2 tissues are mostly unclear. In the current study, the RNA-seq raw data of PM muscle and liver were downloaded from GSE144000, and we constructed the gene coexpression networks of PM muscle and liver to explore the relationships between WB and the 2 tissues using the weighted gene coexpression network analysis (WGCNA) method. Six and 2 gene coexpression modules were significantly correlated with WB in the PM muscle and liver networks, respectively. TGF-beta signaling, Toll-like receptor signaling and mTOR signaling pathways were significantly enriched in the genes within the 6 gene modules of PM muscle network. Meanwhile, mTOR signaling pathway was significantly enriched in the genes within the 2 gene modules of liver network. In the consensus gene coexpression network across the 2 tissues, salmon module (r = -0.5 and p = 0.05) was significantly negatively correlated with WB, in which Toll-like receptor signaling, apoptosis, and autophagy pathways were significantly enriched. The genes related with the 3 pathways, myeloid differentiation primary response 88 (MYD88), interferon regulatory factor 7 (IRF7), mitogen-activated protein kinase 14 (MAPK14), FBJ murine osteosarcoma viral oncogene homolog (FOS), jun proto-oncogene (JUN), caspase-10, unc-51 like autophagy activating kinase 2 (ULK2) and serine/threonine kinase 11 (LKB1), were identified in salmon module. In this current study, we found that the signaling pathways related with cell inflammation, apoptosis and autophagy might influence WB across 2 tissues in broilers.


Subject(s)
Chickens , Gene Regulatory Networks , Liver , Pectoralis Muscles , Poultry Diseases , Animals , Chickens/genetics , Poultry Diseases/genetics , Poultry Diseases/metabolism , Pectoralis Muscles/metabolism , Liver/metabolism , Muscular Diseases/veterinary , Muscular Diseases/genetics , Muscular Diseases/pathology , Signal Transduction , Avian Proteins/genetics , Avian Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL