Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.182
Filtrer
1.
Cells ; 13(14)2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-39056750

RÉSUMÉ

Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.


Sujet(s)
Cardiomyopathies , Mitochondries , Myopathie de Duchenne , Myopathie de Duchenne/complications , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/anatomopathologie , Humains , Cardiomyopathies/thérapie , Cardiomyopathies/métabolisme , Cardiomyopathies/anatomopathologie , Cardiomyopathies/étiologie , Animaux , Mitochondries/métabolisme , Dystrophine/métabolisme , Dystrophine/génétique , Dystrophine/déficit
2.
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)
Dystrophine , Thérapie génétique , Muscles squelettiques , Myopathie de Duchenne , Animaux , Mâle , Souris , Dependovirus/génétique , Modèles animaux de maladie humaine , Dystrophine/génétique , Dystrophine/métabolisme , Techniques de transfert de gènes , Thérapie génétique/méthodes , Vecteurs génétiques , Souris de lignée mdx , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Myocarde/métabolisme , Myocarde/anatomopathologie , Sarcolemme/métabolisme
3.
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
4.
Physiol Rep ; 12(13): e16145, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001580

RÉSUMÉ

The dystrophin protein has well-characterized roles in force transmission and maintaining membrane integrity during muscle contraction. Studies have reported decreased expression of dystrophin in atrophying muscles during wasting conditions, and that restoration of dystrophin can attenuate atrophy, suggesting a role in maintaining muscle mass. Phosphorylation of S3059 within the cysteine-rich region of dystrophin enhances binding between dystrophin and ß-dystroglycan, and mimicking phosphorylation at this site by site-directed mutagenesis attenuates myotube atrophy in vitro. To determine whether dystrophin phosphorylation can attenuate muscle wasting in vivo, CRISPR-Cas9 was used to generate mice with whole body mutations of S3059 to either alanine (DmdS3059A) or glutamate (DmdS3059E), to mimic a loss of, or constitutive phosphorylation of S3059, on all endogenous dystrophin isoforms, respectively. Sciatic nerve transection was performed on these mice to determine whether phosphorylation of dystrophin S3059 could attenuate denervation atrophy. At 14 days post denervation, atrophy of tibialis anterior (TA) but not gastrocnemius or soleus muscles, was partially attenuated in DmdS3059E mice relative to WT mice. Attenuation of atrophy was associated with increased expression of ß-dystroglycan in TA muscles of DmdS3059E mice. Dystrophin S3059 phosphorylation can partially attenuate denervation-induced atrophy, but may have more significant impact in less severe modes of muscle wasting.


Sujet(s)
Dystrophine , Muscles squelettiques , Amyotrophie , Animaux , Phosphorylation , Souris , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Amyotrophie/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/innervation , Muscles squelettiques/anatomopathologie , Dystrophine/métabolisme , Dystrophine/génétique , Mâle , Dénervation musculaire/méthodes , Souris de lignée C57BL
5.
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
6.
Medicina (Kaunas) ; 60(7)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39064489

RÉSUMÉ

Muscular dystrophies (MDs) are a heterogeneous group of diseases of genetic origin characterized by progressive skeletal muscle degeneration and weakness. There are several types of MDs, varying in terms of age of onset, severity, and pattern of the affected muscles. However, all of them worsen over time, and many patients will eventually lose their ability to walk. In addition to skeletal muscle effects, patients with MDs may present cardiac and respiratory disorders, generating complications that could lead to death. Interdisciplinary management is required to improve the surveillance and quality of life of patients with an MD. At present, pharmacological therapy is only available for Duchene muscular dystrophy (DMD)-the most common type of MD-and is mainly based on the use of corticosteroids. Other MDs caused by alterations in dystrophin-associated proteins (DAPs) are less frequent but represent an important group within these diseases. Pharmacological alternatives with clinical potential in patients with MDs and other proteins associated with dystrophin have been scarcely explored. This review focuses on drugs and molecules that have shown beneficial effects, mainly in experimental models involving alterations in DAPs. The mechanisms associated with the effects leading to promising results regarding the recovery or maintenance of muscle strength and reduction in fibrosis in the less-common MDs (i.e., with respect to DMD) are explored, and other therapeutic targets that could contribute to maintaining the homeostasis of muscle fibers, involving different pathways, such as calcium regulation, hypertrophy, and maintenance of satellite cell function, are also examined. It is possible that some of the drugs explored here could be used to affordably improve the muscular function of patients until a definitive treatment for MDs is developed.


Sujet(s)
Dystrophies musculaires , Humains , Dystrophies musculaires/traitement médicamenteux , Dystrophies musculaires/physiopathologie , Dystrophine , Muscles squelettiques/effets des médicaments et des substances chimiques , Muscles squelettiques/physiopathologie , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/physiopathologie , Complexe protéique associé à la dystrophine
7.
Int J Mol Sci ; 25(14)2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39063034

RÉSUMÉ

Duchenne and Becker muscular dystrophies, caused by pathogenic variants in DMD, are the most common inherited neuromuscular conditions in childhood. These diseases follow an X-linked recessive inheritance pattern, and mainly males are affected. The most prevalent pathogenic variants in the DMD gene are copy number variants (CNVs), and most patients achieve their genetic diagnosis through Multiplex Ligation-dependent Probe Amplification (MLPA) or exome sequencing. Here, we investigated a female patient presenting with muscular dystrophy who remained genetically undiagnosed after MLPA and exome sequencing. RNA sequencing (RNAseq) from the patient's muscle biopsy identified an 85% reduction in DMD expression compared to 116 muscle samples included in the cohort. A de novo balanced translocation between chromosome 17 and the X chromosome (t(X;17)(p21.1;q23.2)) disrupting the DMD and BCAS3 genes was identified through trio whole genome sequencing (WGS). The combined analysis of RNAseq and WGS played a crucial role in the detection and characterisation of the disease-causing variant in this patient, who had been undiagnosed for over two decades. This case illustrates the diagnostic odyssey of female DMD patients with complex structural variants that are not detected by current panel or exome sequencing analysis.


Sujet(s)
Chromosomes X humains , Dystrophine , Génomique , Myopathie de Duchenne , Translocation génétique , Humains , Myopathie de Duchenne/génétique , Myopathie de Duchenne/diagnostic , Femelle , Dystrophine/génétique , Chromosomes X humains/génétique , Génomique/méthodes , Variations de nombre de copies de segment d'ADN , , Transcriptome/génétique , Chromosomes humains de la paire 17/génétique
8.
Nature ; 632(8023): 192-200, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39020181

RÉSUMÉ

Gene replacement using adeno-associated virus (AAV) vectors is a promising therapeutic approach for many diseases1,2. However, this therapeutic modality is challenged by the packaging capacity of AAVs (approximately 4.7 kilobases)3, limiting its application for disorders involving large coding sequences, such as Duchenne muscular dystrophy, with a 14 kilobase messenger RNA. Here we developed a new method for expressing large dystrophins by utilizing the protein trans-splicing mechanism mediated by split inteins. We identified several split intein pairs that efficiently join two or three fragments to generate a large midi-dystrophin or the full-length protein. We show that delivery of two or three AAVs into dystrophic mice results in robust expression of large dystrophins and significant physiological improvements compared with micro-dystrophins. Moreover, using the potent myotropic AAVMYO4, we demonstrate that low total doses (2 × 1013 viral genomes per kg) are sufficient to express large dystrophins in striated muscles body-wide with significant physiological corrections in dystrophic mice. Our data show a clear functional superiority of large dystrophins over micro-dystrophins that are being tested in clinical trials. This method could benefit many patients with Duchenne or Becker muscular dystrophy, regardless of genotype, and could be adapted to numerous other disorders caused by mutations in large genes that exceed the AAV capacity.


Sujet(s)
Dystrophine , Thérapie génétique , Intéines , Myopathie de Duchenne , Épissage des protéines , Animaux , Humains , Mâle , Souris , Dependovirus/génétique , Dependovirus/métabolisme , Modèles animaux de maladie humaine , Dystrophine/biosynthèse , Dystrophine/déficit , Dystrophine/génétique , Dystrophine/métabolisme , Thérapie génétique/méthodes , Vecteurs génétiques/génétique , Vecteurs génétiques/métabolisme , Intéines/génétique , Souris de lignée mdx , Muscles squelettiques/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/métabolisme , Épissage des protéines/génétique
9.
J Physiol ; 602(15): 3641-3660, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38980963

RÉSUMÉ

Limited knowledge exists regarding the chronic effect of muscular exercise on muscle function in a murine model of severe Duchenne muscular dystrophy (DMD). Here we determined the effects of 1 month of voluntary wheel running (WR), 1 month of enforced treadmill running (TR) and 1 month of mechanical overloading resulting from the removal of the synergic muscles (OVL) in mice lacking both dystrophin and desmin (DKO). Additionally, we examined the effect of activin receptor administration (AR). DKO mice, displaying severe muscle weakness, atrophy and greater susceptibility to contraction-induced functional loss, were exercised or treated with AR at 1 month of age and in situ force production of lower leg muscle was measured at the age of 2 months. We found that TR and OVL increased absolute maximal force and the rate of force development of the plantaris muscle in DKO mice. In contrast, those of the tibialis anterior (TA) muscle remained unaffected by TR and WR. Furthermore, the effects of TR and OVL on plantaris muscle function in DKO mice closely resembled those in mdx mice, a less severe murine DMD model. AR also improved absolute maximal force and the rate of force development of the TA muscle in DKO mice. In conclusion, exercise training improved plantaris muscle weakness in severely affected dystrophic mice. Consequently, these preclinical results may contribute to fostering further investigations aimed at assessing the potential benefits of exercise for DMD patients, particularly resistance training involving a low number of intense muscle contractions. KEY POINTS: Very little is known about the effects of exercise training in a murine model of severe Duchenne muscular dystrophy (DMD). One reason is that it is feared that chronic muscular exercise, particularly that involving intense muscle contractions, could exacerbate the disease. In DKO mice lacking both dystrophin and desmin, characterized by severe lower leg muscle weakness, atrophy and fragility in comparison to the less severe DMD mdx model, we found that enforced treadmill running improved absolute maximal force of the plantaris muscle, while that of tibialis anterior muscle remained unaffected by both enforced treadmill and voluntary wheel running. Furthermore, mechanical overloading, a non-physiological model of chronic resistance exercise, reversed plantaris muscle weakness. Consequently, our findings may have the potential to alleviate concerns and pave the way for exploring the prescription of endurance and resistance training as a viable therapeutic approach for the treatment of dystrophic patients. Additionally, such interventions may serve in mitigating the pathophysiological mechanisms induced by physical inactivity.


Sujet(s)
Desmine , Dystrophine , Souris knockout , Muscles squelettiques , Conditionnement physique d'animal , Course à pied , Animaux , Dystrophine/génétique , Muscles squelettiques/physiologie , Muscles squelettiques/métabolisme , Souris , Course à pied/physiologie , Desmine/génétique , Desmine/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/physiopathologie , Force musculaire , Mâle , Souris de lignée C57BL , Souris de lignée mdx , Contraction musculaire
10.
Neurobiol Dis ; 199: 106586, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38950712

RÉSUMÉ

OBJECTIVE: The glymphatic system serves as a perivascular pathway that aids in clearing liquid and solute waste from the brain, thereby enhancing neurological function. Disorders in glymphatic drainage contribute to the development of vasogenic edema following cerebral ischemia, although the molecular mechanisms involved remain poorly understood. This study aims to determine whether a deficiency in dystrophin 71 (DP71) leads to aquaporin-4 (AQP4) depolarization, contributing to glymphatic dysfunction in cerebral ischemia and resulting in brain edema. METHODS: A mice model of middle cerebral artery occlusion and reperfusion was used. A fluorescence tracer was injected into the cortex and evaluated glymphatic clearance. To investigate the role of DP71 in maintaining AQP4 polarization, an adeno-associated virus with the astrocyte promoter was used to overexpress Dp71. The expression and distribution of DP71 and AQP4 were analyzed using immunoblotting, immunofluorescence, and co-immunoprecipitation techniques. The behavior ability of mice was evaluated by open field test. Open-access transcriptome sequencing data were used to analyze the functional changes of astrocytes after cerebral ischemia. MG132 was used to inhibit the ubiquitin-proteasome system. The ubiquitination of DP71 was detected by immunoblotting and co-immunoprecipitation. RESULTS: During the vasogenic edema stage following cerebral ischemia, a decline in the efflux of interstitial fluid tracer was observed. DP71 and AQP4 were co-localized and interacted with each other in the perivascular astrocyte endfeet. After cerebral ischemia, there was a notable reduction in DP71 protein expression, accompanied by AQP4 depolarization and proliferation of reactive astrocytes. Increased DP71 expression restored glymphatic drainage and reduced brain edema. AQP4 depolarization, reactive astrocyte proliferation, and the behavior of mice were improved. After cerebral ischemia, DP71 was degraded by ubiquitination, and MG132 inhibited the decrease of DP71 protein level. CONCLUSION: AQP4 depolarization after cerebral ischemia leads to glymphatic clearance disorder and aggravates cerebral edema. DP71 plays a pivotal role in regulating AQP4 polarization and consequently influences glymphatic function. Changes in DP71 expression are associated with the ubiquitin-proteasome system. This study offers a novel perspective on the pathogenesis of brain edema following cerebral ischemia.


Sujet(s)
Aquaporine-4 , Oedème cérébral , Encéphalopathie ischémique , Dystrophine , Système glymphatique , Animaux , Aquaporine-4/métabolisme , Aquaporine-4/génétique , Souris , Système glymphatique/métabolisme , Encéphalopathie ischémique/métabolisme , Encéphalopathie ischémique/anatomopathologie , Oedème cérébral/métabolisme , Dystrophine/métabolisme , Dystrophine/déficit , Mâle , Astrocytes/métabolisme , Souris de lignée C57BL , Infarctus du territoire de l'artère cérébrale moyenne/métabolisme
11.
Nat Commun ; 15(1): 4935, 2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38858388

RÉSUMÉ

Cell polarity mechanisms allow the formation of specialized membrane domains with unique protein compositions, signalling properties, and functional characteristics. By analyzing the localization of potassium channels and proteins belonging to the dystrophin-associated protein complex, we reveal the existence of distinct planar-polarized membrane compartments at the surface of C. elegans muscle cells. We find that muscle polarity is controlled by a non-canonical Wnt signalling cascade involving the ligand EGL-20/Wnt, the receptor CAM-1/Ror, and the intracellular effector DSH-1/Dishevelled. Interestingly, classical planar cell polarity proteins are not required for this process. Using time-resolved protein degradation, we demonstrate that -while it is essentially in place by the end of embryogenesis- muscle polarity is a dynamic state, requiring continued presence of DSH-1 throughout post-embryonic life. Our results reveal the unsuspected complexity of the C. elegans muscle membrane and establish a genetically tractable model system to study cellular polarity and membrane compartmentalization in vivo.


Sujet(s)
Protéines de Caenorhabditis elegans , Caenorhabditis elegans , Polarité de la cellule , Dystrophine , Muscles , Voie de signalisation Wnt , Animaux , Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/métabolisme , Protéines de Caenorhabditis elegans/génétique , Dystrophine/métabolisme , Dystrophine/génétique , Muscles/métabolisme , Protéines Dishevelled/métabolisme , Protéines Dishevelled/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Membrane cellulaire/métabolisme , Complexe protéique associé à la dystrophine/métabolisme , Complexe protéique associé à la dystrophine/génétique , Protéines de type Wingless/métabolisme , Transduction du signal
12.
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
13.
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
14.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38892308

RÉSUMÉ

Sarcospan (SSPN) is a 25-kDa transmembrane protein that is broadly expressed at the cell surface of many tissues, including, but not limited to, the myofibers from skeletal and smooth muscles, cardiomyocytes, adipocytes, kidney epithelial cells, and neurons. SSPN is a core component of the dystrophin-glycoprotein complex (DGC) that links the intracellular actin cytoskeleton with the extracellular matrix. It is also associated with integrin α7ß1, the predominant integrin expressed in skeletal muscle. As a tetraspanin-like protein with four transmembrane spanning domains, SSPN functions as a scaffold to facilitate protein-protein interactions at the cell membrane. Duchenne muscular dystrophy, Becker muscular dystrophy, and X-linked dilated cardiomyopathy are caused by the loss of dystrophin at the muscle cell surface and a concomitant loss of the entire DGC, including SSPN. SSPN overexpression ameliorates Duchenne muscular dystrophy in the mdx murine model, which supports SSPN being a viable therapeutic target. Other rescue studies support SSPN as a biomarker for the proper assembly and membrane expression of the DGC. Highly specific and robust antibodies to SSPN are needed for basic research on the molecular mechanisms of SSPN rescue, pre-clinical studies, and biomarker evaluations in human samples. The development of SSPN antibodies is challenged by the presence of its four transmembrane domains and limited antigenic epitopes. To address the significant barrier presented by limited commercially available antibodies, we aimed to generate a panel of robust SSPN-specific antibodies that can serve as a resource for the research community. We created antibodies to three SSPN protein epitopes, including the intracellular N- and C-termini as well as the large extracellular loop (LEL) between transmembrane domains 3 and 4. We developed a panel of rabbit antibodies (poly- and monoclonal) against an N-terminal peptide fragment of SSPN. We used several assays to show that the rabbit antibodies recognize mouse SSPN with a high functional affinity and specificity. We developed mouse monoclonal antibodies against the C-terminal peptide and the large extracellular loop of human SSPN. These antibodies are superior to commercially available antibodies and outperform them in various applications, including immunoblotting, indirect immunofluorescence analysis, immunoprecipitation, and an ELISA. These newly developed antibodies will significantly improve the quality and ease of SSPN detection for basic and translational research.


Sujet(s)
Protéines membranaires , , Animaux , Humains , Souris , Dystrophine/métabolisme , Dystrophine/immunologie , Dystrophine/génétique , Intégrines/métabolisme , Intégrines/immunologie , Protéines membranaires/immunologie , Protéines membranaires/métabolisme , Myopathie de Duchenne/immunologie , Myopathie de Duchenne/métabolisme
15.
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
16.
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
17.
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
18.
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
19.
Biomolecules ; 14(5)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38785982

RÉSUMÉ

Chimerism-based strategies represent a pioneering concept which has led to groundbreaking advancements in regenerative medicine and transplantation. This new approach offers therapeutic potential for the treatment of various diseases, including inherited disorders. The ongoing studies on chimeric cells prompted the development of Dystrophin-Expressing Chimeric (DEC) cells which were introduced as a potential therapy for Duchenne Muscular Dystrophy (DMD). DMD is a genetic condition that leads to premature death in adolescent boys and remains incurable with current methods. DEC therapy, created via the fusion of human myoblasts derived from normal and DMD-affected donors, has proven to be safe and efficacious when tested in experimental models of DMD after systemic-intraosseous administration. These studies confirmed increased dystrophin expression, which correlated with functional and morphological improvements in DMD-affected muscles, including cardiac, respiratory, and skeletal muscles. Furthermore, the application of DEC therapy in a clinical study confirmed its long-term safety and efficacy in DMD patients. This review summarizes the development of chimeric cell technology tested in preclinical models and clinical studies, highlighting the potential of DEC therapy in muscle regeneration and repair, and introduces chimeric cell-based therapies as a promising, novel approach for muscle regeneration and the treatment of DMD and other neuromuscular disorders.


Sujet(s)
Thérapie cellulaire et tissulaire , Dystrophine , Muscles squelettiques , Myopathie de Duchenne , Régénération , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/génétique , Humains , Animaux , Thérapie cellulaire et tissulaire/méthodes , Dystrophine/génétique , Dystrophine/métabolisme , Myoblastes/métabolisme
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE