Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.057
Filtrer
1.
FASEB J ; 38(14): e23771, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-38989564

RÉSUMÉ

DUX4 has been widely reported in facioscapulohumeral muscular dystrophy, but its role in Duchenne muscular dystrophy (DMD) is unclear. Dux is the mouse paralog of DUX4. In Dux-/- mdx mice, forelimb grip strength test and treadmill test were performed, and extensor digitorum longus (EDL) contraction properties were measured to assess skeletal muscle function. Pathological changes in mice were determined by serum CK and LDH levels and muscle Masson staining. Inflammatory factors, oxidative stress, and mitochondrial function indicators were detected using kits. Primary muscle satellite cells were isolated, and the antioxidant molecule Nrf2 was detected. MTT assay and Edu assay were used to evaluate proliferation and TUNEL assay for cell death. The results show that the deletion of Dux enhanced forelimb grip strength and EDL contractility, prolonged running time and distance in mdx mice. Deleting Dux also attenuated muscle fibrosis, inflammation, oxidative stress, and mitochondrial dysfunction in mdx mice. Furthermore, Dux deficiency promoted proliferation and survival of muscle satellite cells by increasing Nrf2 levels in mdx mice.


Sujet(s)
Protéines à homéodomaine , Souris de lignée mdx , Myopathie de Duchenne , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Souris , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Mâle , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Cellules satellites du muscle squelettique/métabolisme , Souris de lignée C57BL , Souris knockout , Délétion de gène
2.
Nat Commun ; 15(1): 5927, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-39009678

RÉSUMÉ

Duchenne muscular dystrophy (DMD) affecting 1 in 3500-5000 live male newborns is the frequently fatal genetic disease resulted from various mutations in DMD gene encoding dystrophin protein. About 70% of DMD-causing mutations are exon deletion leading to frameshift of open reading frame and dystrophin deficiency. To facilitate translating human DMD-targeting CRISPR therapeutics into patients, we herein establish a genetically humanized mouse model of DMD by replacing exon 50 and 51 of mouse Dmd gene with human exon 50 sequence. This humanized mouse model recapitulats patient's DMD phenotypes of dystrophin deficiency and muscle dysfunction. Furthermore, we target splicing sites in human exon 50 with adenine base editor to induce exon skipping and robustly restored dystrophin expression in heart, tibialis anterior and diaphragm muscles. Importantly, systemic delivery of base editor via adeno-associated virus in the humanized male mouse model improves the muscle function of DMD mice to the similar level of wildtype ones, indicating the therapeutic efficacy of base editing strategy in treating most of DMD types with exon deletion or point mutations via exon-skipping induction.


Sujet(s)
Adénine , Systèmes CRISPR-Cas , Modèles animaux de maladie humaine , Dystrophine , Exons , Édition de gène , Myopathie de Duchenne , Animaux , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Dystrophine/génétique , Dystrophine/métabolisme , Exons/génétique , Humains , Mâle , Édition de gène/méthodes , Souris , Adénine/métabolisme , Muscles squelettiques/métabolisme , Dependovirus/génétique , Thérapie génétique/méthodes
3.
Nat Commun ; 15(1): 6141, 2024 Jul 21.
Article de Anglais | MEDLINE | ID: mdl-39034316

RÉSUMÉ

Current gene therapy for Duchenne muscular dystrophy (DMD) utilizes adeno-associated virus (AAV) to deliver micro-dystrophin (µDys), which does not provide full protection for striated muscles as it lacks many important functional domains of full-length (FL) dystrophin. Here we develop a triple vector system to deliver FL-dystrophin into skeletal and cardiac muscles. We split FL-dystrophin into three fragments linked to two orthogonal pairs of split intein, allowing efficient assembly of FL-dystrophin. The three fragments packaged in myotropic AAV (MyoAAV4A) restore FL-dystrophin expression in both skeletal and cardiac muscles in male mdx4cv mice. Dystrophin-glycoprotein complex components are also restored at the sarcolemma of dystrophic muscles. MyoAAV4A-delivered FL-dystrophin significantly improves muscle histopathology, contractility, and overall strength comparable to µDys, but unlike µDys, it also restores defective cavin 4 localization and associated signaling in mdx4cv heart. Therefore, our data support the feasibility of a mutation-independent FL-dystrophin gene therapy for DMD, warranting further clinical development.


Sujet(s)
Dependovirus , Dystrophine , Thérapie génétique , Vecteurs génétiques , Souris de lignée mdx , Muscles squelettiques , Myopathie de Duchenne , Animaux , Dystrophine/génétique , Dystrophine/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/anatomopathologie , Thérapie génétique/méthodes , Dependovirus/génétique , Souris , Mâle , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Vecteurs génétiques/génétique , Vecteurs génétiques/administration et posologie , Modèles animaux de maladie humaine , Myocarde/métabolisme , Myocarde/anatomopathologie , Sarcolemme/métabolisme , Humains , Techniques de transfert de gènes
4.
Yi Chuan ; 46(7): 570-580, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39016090

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is a severe X-linked recessive genetic disorder caused by mutations in the DMD gene, which leads to a deficiency of the dystrophin protein. The main mutation types of this gene include exon deletions and duplications, point mutations, and insertions. These mutations disrupt the normal expression of dystrophin, ultimately leading to the disease. In this study, we reported a case of DMD caused by an insertion mutation in exon 59 (E59) of the DMD gene. The affected child exhibited significant abnormalities in related biochemical markers, early symptoms of DMD, and multiple gray hair. His mother and sister were carriers with slightly abnormal biochemical markers. The mother had mild clinical symptoms, while the sister had no clinical symptoms. Other family members were genetically and physically normal. Sequencing and sequence alignment revealed that the inserted fragment was an Alu element from the AluYa5 subfamily. This insertion produced two stop codons and a polyadenylate (polyA) tail. To understand the impact of this insertion on the DMD gene and its association with clinical symptoms, exonic splicing enhancer (ESE) prediction indicated that the insertion did not affect the splicing of E59. Therefore, we speculated that the insertion sequence would be present in the mRNA sequence of the DMD gene. The two stop codons and polyA tail likely terminate translation, preventing the production of functional dystrophin protein, which may be the mechanism leading to DMD. In addition to typical DMD symptoms, the child also exhibited premature graying of hair. This study reports, for the first time, a case of DMD caused by the insertion of an Alu element into the coding region of the DMD gene. This finding provides clues for studying gene mutations induced by Alu sequence insertion and expands the understanding of DMD gene mutations.


Sujet(s)
Séquences Alu , Dystrophine , Myopathie de Duchenne , Mutagenèse par insertion , Myopathie de Duchenne/génétique , Humains , Séquences Alu/génétique , Dystrophine/génétique , Mâle , Séquence nucléotidique , Poils/métabolisme , Femelle , Exons/génétique , Enfant , Données de séquences moléculaires
5.
PLoS Genet ; 20(6): e1010935, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38875306

RÉSUMÉ

Gene regulatory networks that act upstream of skeletal muscle fate determinants are distinct in different anatomical locations. Despite recent efforts, a clear understanding of the cascade of events underlying the emergence and maintenance of the stem cell pool in specific muscle groups remains unresolved and debated. Here, we invalidated Pitx2 with multiple Cre-driver mice prenatally, postnatally, and during lineage progression. We showed that this gene becomes progressively dispensable for specification and maintenance of the muscle stem (MuSC) cell pool in extraocular muscles (EOMs) despite being, together with Myf5, a major upstream regulator during early development. Moreover, constitutive inactivation of Pax7 postnatally led to a greater loss of MuSCs in the EOMs compared to the limb. Thus, we propose a relay between Pitx2, Myf5 and Pax7 for EOM stem cell maintenance. We demonstrate also that MuSCs in the EOMs adopt a quiescent state earlier that those in limb muscles and do not spontaneously proliferate in the adult, yet EOMs have a significantly higher content of Pax7+ MuSCs per area pre- and post-natally. Finally, while limb MuSCs proliferate in the mdx mouse model for Duchenne muscular dystrophy, significantly less MuSCs were present in the EOMs of the mdx mouse model compared to controls, and they were not proliferative. Overall, our study provides a comprehensive in vivo characterisation of MuSC heterogeneity along the body axis and brings further insights into the unusual sparing of EOMs during muscular dystrophy.


Sujet(s)
, Protéines à homéodomaine , Facteur-5 de régulation myogène , Muscles oculomoteurs , Facteur de transcription PAX7 , Facteurs de transcription , Animaux , Humains , Souris , Différenciation cellulaire/génétique , Lignage cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes au cours du développement , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Souris de lignée mdx , Développement musculaire/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/croissance et développement , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Facteur-5 de régulation myogène/génétique , Facteur-5 de régulation myogène/métabolisme , Muscles oculomoteurs/métabolisme , Facteur de transcription PAX7/métabolisme , Facteur de transcription PAX7/génétique , Cellules souches/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
6.
Genes Brain Behav ; 23(3): e12895, 2024 06.
Article de Anglais | MEDLINE | ID: mdl-38837620

RÉSUMÉ

Duchenne muscular dystrophy is a severe neuromuscular disorder that is caused by mutations in the DMD gene, resulting in a disruption of dystrophin production. Next to dystrophin expression in the muscle, different isoforms of the protein are also expressed in the brain and lack of these isoforms leads to cognitive and behavioral deficits in patients. It remains unclear how the loss of the shorter dystrophin isoform Dp140 affects these processes. Using a variety of behavioral tests, we found that mdx and mdx4cv mice (which lack Dp427 or Dp427 + Dp140, respectively) exhibit similar deficits in working memory, movement patterns and blood-brain barrier integrity. Neither model showed deficits in spatial learning and memory, learning flexibility, anxiety or spontaneous behavior, nor did we observe differences in aquaporin 4 and glial fibrillary acidic protein. These results indicate that in contrast to Dp427, Dp140 does not play a crucial role in processes of learning, memory and spontaneous behavior.


Sujet(s)
Barrière hémato-encéphalique , Dystrophine , Myopathie de Duchenne , Animaux , Souris , Barrière hémato-encéphalique/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/physiopathologie , Dystrophine/génétique , Dystrophine/métabolisme , Mâle , Souris de lignée mdx , Souris de lignée C57BL , Aquaporine-4/génétique , Aquaporine-4/métabolisme , Mémoire à court terme , Mémoire
8.
FASEB J ; 38(11): e23718, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38847487

RÉSUMÉ

Female carriers of a Duchenne muscular dystrophy (DMD) gene mutation manifest exercise intolerance and metabolic anomalies that may be exacerbated following menopause due to the loss of estrogen, a known regulator of skeletal muscle function and metabolism. Here, we studied the impact of estrogen depletion (via ovariectomy) on exercise tolerance and muscle mitochondrial metabolism in female mdx mice and the potential of estrogen replacement therapy (using estradiol) to protect against functional and metabolic perturbations. We also investigated the effect of estrogen depletion, and replacement, on the skeletal muscle proteome through an untargeted proteomic approach with TMT-labelling. Our study confirms that loss of estrogen in female mdx mice reduces exercise capacity, tricarboxylic acid cycle intermediates, and citrate synthase activity but that these deficits are offset through estrogen replacement therapy. Furthermore, ovariectomy downregulated protein expression of RNA-binding motif factor 20 (Rbm20), a critical regulator of sarcomeric and muscle homeostasis gene splicing, which impacted pathways involving ribosomal and mitochondrial translation. Estrogen replacement modulated Rbm20 protein expression and promoted metabolic processes and the upregulation of proteins involved in mitochondrial dynamics and metabolism. Our data suggest that estrogen mitigates dystrophinopathic features in female mdx mice and that estrogen replacement may be a potential therapy for post-menopausal DMD carriers.


Sujet(s)
Oestrogènes , Souris de lignée mdx , Muscles squelettiques , Protéines de liaison à l'ARN , Animaux , Femelle , Souris , Oestrogènes/métabolisme , Oestrogènes/pharmacologie , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/génétique , Souris de lignée C57BL , Ovariectomie , Mitochondries/métabolisme , Mitochondries du muscle/métabolisme , Mitochondries du muscle/effets des médicaments et des substances chimiques
9.
Skelet Muscle ; 14(1): 13, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38867250

RÉSUMÉ

BACKGROUND: Adult muscle-resident myogenic stem cells, satellite cells (SCs), that play non-redundant role in muscle regeneration, are intrinsically impaired in Duchenne muscular dystrophy (DMD). Previously we revealed that dystrophic SCs express low level of anti-inflammatory and anti-oxidative heme oxygenase-1 (HO-1, HMOX1). Here we assess whether targeted induction of HMOX1 affect SC function and alleviates hallmark symptoms of DMD. METHODS: We generated double-transgenic mouse model (mdx;HMOX1Pax7Ind) that allows tamoxifen (TX)-inducible HMOX1 expression in Pax7 positive cells of dystrophic muscles. Mdx;HMOX1Pax7Ind and control mdx mice were subjected to 5-day TX injections (75 mg/kg b.w.) followed by acute exercise protocol with high-speed treadmill (12 m/min, 45 min) and downhill running to worsen skeletal muscle phenotype and reveal immediate effects of HO-1 on muscle pathology and SC function. RESULTS: HMOX1 induction caused a drop in SC pool in mdx;HMOX1Pax7Ind mice (vs. mdx counterparts), while not exaggerating the effect of physical exercise. Upon physical exercise, the proliferation of SCs and activated CD34- SC subpopulation, was impaired in mdx mice, an effect that was reversed in mdx;HMOX1Pax7Ind mice, however, both in vehicle- and TX-treated animals. This corresponded to the pattern of HO-1 expression in skeletal muscles. At the tissue level, necrotic events of selective skeletal muscles of mdx mice and associated increase in circulating levels of muscle damage markers were blunted in HO-1 transgenic animals which showed also anti-inflammatory cytokine profile (vs. mdx). CONCLUSIONS: Targeted expression of HMOX1 plays protective role in DMD and alleviates dystrophic muscle pathology.


Sujet(s)
Heme oxygenase-1 , Souris de lignée mdx , Souris transgéniques , Muscles squelettiques , Myopathie de Duchenne , Cellules satellites du muscle squelettique , Animaux , Heme oxygenase-1/génétique , Heme oxygenase-1/métabolisme , Cellules satellites du muscle squelettique/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Souris , Facteur de transcription PAX7/génétique , Facteur de transcription PAX7/métabolisme , Mâle , Souris de lignée C57BL , Conditionnement physique d'animal , Protéines membranaires
10.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892293

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is an X-linked progressive disorder associated with muscle wasting and degeneration. The disease is caused by mutations in the gene that encodes dystrophin, a protein that links the cytoskeleton with cell membrane proteins. The current treatment methods aim to relieve the symptoms of the disease or partially rescue muscle functionality. However, they are insufficient to suppress disease progression. In recent years, studies have uncovered an important role for non-coding RNAs (ncRNAs) in regulating the progression of numerous diseases. ncRNAs, such as micro-RNAs (miRNAs), bind to their target messenger RNAs (mRNAs) to suppress translation. Understanding the mechanisms involving dysregulated miRNAs can improve diagnosis and suggest novel treatment methods for patients with DMD. This review presents the available evidence on the role of altered expression of miRNAs in the pathogenesis of DMD. We discuss the involvement of these molecules in the processes associated with muscle physiology and DMD-associated cardiomyopathy.


Sujet(s)
microARN , Myopathie de Duchenne , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Humains , microARN/génétique , microARN/métabolisme , Animaux , Dystrophine/génétique , Dystrophine/métabolisme , Régulation de l'expression des gènes , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie
11.
Cells ; 13(11)2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38891104

RÉSUMÉ

Mutations in the DMD gene cause fatal Duchenne Muscular Dystrophy (DMD). An attractive therapeutic approach is autologous cell transplantation utilizing myogenic progenitors derived from induced pluripotent stem cells (iPSCs). Given that a significant number of DMD mutations occur between exons 45 and 55, we developed a gene knock-in approach to correct any mutations downstream of exon 44. We applied this approach to two DMD patient-specific iPSC lines carrying mutations in exons 45 and 51 and confirmed mini-DYSTROPHIN (mini-DYS) protein expression in corrected myotubes by western blot and immunofluorescence staining. Transplantation of gene-edited DMD iPSC-derived myogenic progenitors into NSG/mdx4Cv mice produced donor-derived myofibers, as shown by the dual expression of human DYSTROPHIN and LAMIN A/C. These findings further provide proof-of-concept for the use of programmable nucleases for the development of autologous iPSC-based therapy for muscular dystrophies.


Sujet(s)
Dystrophine , Édition de gène , Cellules souches pluripotentes induites , Myopathie de Duchenne , Mutation , Animaux , Humains , Souris , Systèmes CRISPR-Cas/génétique , Dystrophine/génétique , Dystrophine/métabolisme , Exons/génétique , Édition de gène/méthodes , Cellules souches pluripotentes induites/métabolisme , Fibres musculaires squelettiques/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/anatomopathologie
12.
Neurogenetics ; 25(3): 201-213, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38850354

RÉSUMÉ

Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are X-linked recessive allelic muscle diseases caused by dystrophin gene mutations. Eight hundred thirty-seven patients admitted between 1997 and 2022 were included in the study. Two hundred twenty patients were analyzed by multiplex PCR (mPCR) alone. Five hundred ninety-five patients were investigated by multiplex ligation-dependent probe amplification (MLPA), and 54 patients were examined by sequencing. Deletion was detected in 60% (132/220) of the cases in the mPCR group only and in 58.3% (347/595) of the cases with MLPA analysis. The rates of deletion and duplication were 87.7% and 12.3%, respectively, in the MLPA analysis. Single exon deletions were the most common mutation type. The introns 43-55 (81.8%) and exons 2-21 (13.1%) regions were detected as hot spots in deletions. It was determined that 89% of the mutations were suitable for exon skipping therapy. The reading frame rule did not hold in 7.6% of D/BMD cases (17/224). We detected twenty-five pathogenic/likely pathogenic variants in sequencing, five of which were novel variants. Nonsense mutation was the most common small mutation (44%). 21% of DMD patients were familial. We detected germline mosaicism in four families (4.3%) in the large rearrangement group and one gonosomal mosaicism in a family with a nonsense mutation. This is the largest study examining genotype and phenotype data in Turkish D/BMD families investigated by MLPA analysis. The reading frame hypothesis is not valid in all cases. Sharing the genotype and phenotype characteristics of these cases in the literature will shed light on the molecular structure of DMD and guide gene therapy research. In genetic counseling, carrier screening in the family and possible gonadal mosaicism should be emphasized.


Sujet(s)
Dystrophine , Exons , Myopathie de Duchenne , Phénotype , Humains , Myopathie de Duchenne/génétique , Turquie , Mâle , Dystrophine/génétique , Enfant , Femelle , Adolescent , Enfant d'âge préscolaire , Exons/génétique , Études d'associations génétiques/méthodes , Mutation , Adulte , Génotype , Jeune adulte , Réaction de polymérisation en chaine multiplex
14.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892298

RÉSUMÉ

Periostin, a multifunctional 90 kDa protein, plays a pivotal role in the pathogenesis of fibrosis across various tissues, including skeletal muscle. It operates within the transforming growth factor beta 1 (Tgf-ß1) signalling pathway and is upregulated in fibrotic tissue. Alternative splicing of Periostin's C-terminal region leads to six protein-coding isoforms. This study aimed to elucidate the contribution of the isoforms containing the amino acids encoded by exon 17 (e17+ Periostin) to skeletal muscle fibrosis and investigate the therapeutic potential of manipulating exon 17 splicing. We identified distinct structural differences between e17+ Periostin isoforms, affecting their interaction with key fibrotic proteins, including Tgf-ß1 and integrin alpha V. In vitro mouse fibroblast experimentation confirmed the TGF-ß1-induced upregulation of e17+ Periostin mRNA, mitigated by an antisense approach that induces the skipping of exon 17 of the Postn gene. Subsequent in vivo studies in the D2.mdx mouse model of Duchenne muscular dystrophy (DMD) demonstrated that our antisense treatment effectively reduced e17+ Periostin mRNA expression, which coincided with reduced full-length Periostin protein expression and collagen accumulation. The grip strength of the treated mice was rescued to the wild-type level. These results suggest a pivotal role of e17+ Periostin isoforms in the fibrotic pathology of skeletal muscle and highlight the potential of targeted exon skipping strategies as a promising therapeutic approach for mitigating fibrosis-associated complications.


Sujet(s)
Épissage alternatif , Molécules d'adhérence cellulaire , Exons , Fibrose , Souris de lignée mdx , Oligonucléotides antisens , Animaux , Molécules d'adhérence cellulaire/génétique , Molécules d'adhérence cellulaire/métabolisme , Souris , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/génétique , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Facteur de croissance transformant bêta-1/métabolisme , Facteur de croissance transformant bêta-1/génétique , Fibroblastes/métabolisme , Modèles animaux de maladie humaine , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Mâle
15.
Ann Clin Transl Neurol ; 11(6): 1456-1464, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38693632

RÉSUMÉ

OBJECTIVE: Duchenne and Becker muscular dystrophies (DMD and BMD) are dystrophinopathies caused by variants in DMD gene, resulting in reduced or absent dystrophin. These conditions, characterized by muscle weakness, also manifest central nervous system (CNS) comorbidities due to dystrophin expression in the CNS. Prior studies have indicated a higher prevalence of epilepsy in individuals with dystrophinopathy compared to the general population. Our research aimed to investigate epilepsy prevalence in dystrophinopathies and characterize associated electroencephalograms (EEGs) and seizures. METHODS: We reviewed 416 individuals with dystrophinopathy, followed up at three centers between 2010 and 2023, to investigate the lifetime epilepsy prevalence and characterize EEGs and seizures in those individuals diagnosed with epilepsy. Associations between epilepsy and type of dystrophinopathy, genotype, and cognitive involvement were studied. RESULTS: Our study revealed a higher epilepsy prevalence than the general population (1.4%; 95% confidence interval: 0.7-3.2%), but notably lower than previously reported in smaller dystrophinopathy cohorts. No significant differences were found in epilepsy prevalence between DMD and BMD or based on underlying genotypes. Cognitive impairment was not found to be linked to higher epilepsy rates. The most prevalent epilepsy types in dystrophinopathies resembled those observed in the broader pediatric population, with most individuals effectively controlled through monotherapy. INTERPRETATION: The actual epilepsy prevalence in dystrophinopathies may be markedly lower than previously estimated, possibly half or even less. Our study provides valuable insights into the epilepsy landscape in individuals with dystrophinopathy, impacting medical care, especially for those with concurrent epilepsy.


Sujet(s)
Épilepsie , Myopathie de Duchenne , Humains , Myopathie de Duchenne/épidémiologie , Myopathie de Duchenne/complications , Myopathie de Duchenne/génétique , Mâle , Épilepsie/épidémiologie , Épilepsie/étiologie , Adolescent , Femelle , Adulte , Jeune adulte , Enfant , Prévalence , Adulte d'âge moyen , Enfant d'âge préscolaire , Électroencéphalographie , Comorbidité , Dystrophine/génétique
16.
Neuromuscul Disord ; 39: 24-29, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38714145

RÉSUMÉ

Structural variants (SVs) are infrequently observed in Duchenne muscular dystrophy (DMD), a condition mainly marked by deletions and point mutations in the DMD gene. SVs in DMD remain difficult to reliably detect due to the limited SV-detection capacity of conventionally used short-read sequencing technology. Herein, we present a family, a boy and his mother, with clinical signs of muscular dystrophy, elevated creatinine kinase levels, and intellectual disability. A muscle biopsy from the boy showed dystrophin deficiency. Routine molecular techniques failed to detect abnormalities in the DMD gene, however, dystrophin mRNA transcripts analysis revealed an absence of exons 59 to 79. Subsequent long-read whole-genome sequencing identified a rare complex structural variant, a 77 kb novel intragenic inversion, and a balanced translocation t(X;1)(p21.2;p13.3) rearrangement within the DMD gene, expanding the genetic spectrum of dystrophinopathy. Our findings suggested that SVs should be considered in cases where conventional molecular techniques fail to identify pathogenic variants.


Sujet(s)
Dystrophine , Myopathie de Duchenne , Translocation génétique , Myopathie de Duchenne/génétique , Humains , Mâle , Dystrophine/génétique , Femelle , Inversion chromosomique/génétique , Adulte , Enfant
17.
Respir Physiol Neurobiol ; 326: 104282, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38782084

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is the most common X-linked disease. DMD is caused by a lack of dystrophin, a critical structural protein in striated muscle. Dystrophin deficiency leads to inflammation, fibrosis, and muscle atrophy. Boys with DMD have progressive muscle weakness within the diaphragm that results in respiratory failure in the 2nd or 3rd decade of life. The most common DMD mouse model - the mdx mouse - is not sufficient for evaluating genetic medicines that specifically target the human DMD (hDMD) gene sequence. Therefore, a novel transgenic mouse carrying the hDMD gene with an exon 52 deletion was created (hDMDΔ52;mdx). We characterized the respiratory function and pathology in this model using whole body plethysmography, histology, and immunohistochemistry. At 6-months-old, hDMDΔ52;mdx mice have reduced maximal respiration, neuromuscular junction pathology, and fibrosis throughout the diaphragm, which worsens at 12-months-old. In conclusion, the hDMDΔ52;mdx exhibits moderate respiratory pathology, and serves as a relevant animal model to study the impact of novel genetic therapies, including gene editing, on respiratory function.


Sujet(s)
Modèles animaux de maladie humaine , Souris transgéniques , Myopathie de Duchenne , Animaux , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Myopathie de Duchenne/physiopathologie , Souris , Humains , Mâle , Dystrophine/génétique , Dystrophine/déficit , Souris de lignée mdx , Muscle diaphragme/physiopathologie , Muscle diaphragme/anatomopathologie , Insuffisance respiratoire/étiologie , Jonction neuromusculaire/anatomopathologie , Jonction neuromusculaire/métabolisme , Souris de lignée C57BL
18.
JAMA ; 331(20): 1705-1706, 2024 05 28.
Article de Anglais | MEDLINE | ID: mdl-38691382

RÉSUMÉ

This Viewpoint examines the appropriateness of FDA accelerated approval of novel gene therapies to treat boys with Duchenne muscular dystrophy following clinical trials with surrogate outcomes that did not demonstrate net benefits.


Sujet(s)
Thérapie génétique , Myopathie de Duchenne , Food and Drug Administration (USA) , Humains , Agrément de médicaments , Dystrophine/génétique , Thérapie génétique/effets indésirables , Thérapie génétique/méthodes , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/génétique , États-Unis , Mâle , Enfant d'âge préscolaire , Enfant , Essais cliniques comme sujet
20.
JCI Insight ; 9(11)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38713520

RÉSUMÉ

Clinical trials delivering high doses of adeno-associated viruses (AAVs) expressing truncated dystrophin molecules (microdystrophins) are underway for Duchenne muscular dystrophy (DMD). We examined the efficiency and efficacy of this strategy with 4 microdystrophin constructs (3 in clinical trials and a variant of the largest clinical construct), in a severe mouse model of DMD, using AAV doses comparable with those in clinical trials. We achieved high levels of microdystrophin expression in striated muscles with cardiac expression approximately 10-fold higher than that observed in skeletal muscle. Significant, albeit incomplete, correction of skeletal muscle disease was observed. Surprisingly, a lethal acceleration of cardiac disease occurred with 2 of the microdystrophins. The detrimental cardiac effect appears to be caused by variable competition (dependent on microdystrophin design and expression level) between microdystrophin and utrophin at the cardiomyocyte membrane. There may also be a contribution from an overloading of protein degradation. The significance of these observations for patients currently being treated with AAV-microdystrophin therapies is unclear since the levels of expression being achieved in the DMD hearts are unknown. However, these findings suggest that microdystrophin treatments need to avoid excessively high levels of expression in the heart and that cardiac function should be carefully monitored in these patients.


Sujet(s)
Thérapie génétique , Myopathie de Duchenne , Animaux , Humains , Mâle , Souris , Dependovirus/génétique , Modèles animaux de maladie humaine , Dystrophine/génétique , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Souris de lignée mdx , Muscles squelettiques/métabolisme , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myocytes cardiaques/métabolisme , Utrophine/génétique , Utrophine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE