Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.028
Filtrer
1.
Sleep Med Clin ; 19(3): 473-483, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39095144

RÉSUMÉ

Spinal muscular atrophy (SMA) and Duchenne muscular dystrophy (DMD) are neuromuscular disorders that affect muscular function. The most common causes of morbidity and mortality are respiratory complications, including restrictive lung disease, ineffective cough, and sleep-disordered breathing. The paradigm of care is changing as new disease-modifying therapies are altering disease trajectory, outcomes, expectations, as well as patient and caregiver experiences. This article provides an overview on therapeutic advances for SMA and DMD in the last 10 years, with a focus on the effects of disease-modifying therapies on respiratory function.


Sujet(s)
Myopathie de Duchenne , Humains , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/physiopathologie , Myopathie de Duchenne/complications , Amyotrophie spinale/traitement médicamenteux , Amyotrophie spinale/physiopathologie , Maladies neuromusculaires/traitement médicamenteux , Maladies neuromusculaires/physiopathologie , Maladies neuromusculaires/thérapie
2.
Mol Biomed ; 5(1): 31, 2024 08 09.
Article de Anglais | MEDLINE | ID: mdl-39117956

RÉSUMÉ

Sestrin2 (Sesn2) has been previously confirmed to be a stress-response molecule. However, the influence of Sesn2 on myogenic differentiation remains elusive. This study was conducted to analyze the role of Sesn2 in the myogenic differentiation of C2C12 myoblasts and related aspects in mdx mice, an animal model of Duchenne muscular dystrophy (DMD). Our results showed that knockdown of Sesn2 reduced the myogenic differentiation capacity of C2C12 myoblasts. Predictive analysis from two databases suggested that miR-182-5p is a potential regulator of Sesn2. Further experimental validation revealed that overexpression of miR-182-5p decreased both the protein and mRNA levels of Sesn2 and inhibited myogenesis of C2C12 myoblasts. These findings suggest that miR-182-5p negatively regulates myogenesis by repressing Sesn2 expression. Extending to an in vivo model of DMD, knockdown of Sesn2 led to decreased Myogenin (Myog) expression and increased Pax7 expression, while its overexpression upregulated Myog levels and enhanced the proportion of slow-switch myofibers. These findings indicate the crucial role of Sesn2 in promoting myogenic differentiation and skeletal muscle regeneration, providing potential therapeutic targets for muscular dystrophy.


Sujet(s)
Différenciation cellulaire , Souris de lignée mdx , microARN , Développement musculaire , Myoblastes , Myogénine , Animaux , Myoblastes/métabolisme , Souris , Développement musculaire/physiologie , Développement musculaire/génétique , Lignée cellulaire , Myogénine/génétique , Myogénine/métabolisme , microARN/génétique , microARN/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Techniques de knock-down de gènes , Facteur de transcription PAX7/métabolisme , Facteur de transcription PAX7/génétique , Régulation de l'expression des gènes , Sestrines
3.
Life Sci Alliance ; 7(11)2024 Nov.
Article de Anglais | MEDLINE | ID: mdl-39117454

RÉSUMÉ

Structural variants (SVs) of unknown significance are great challenges for prenatal risk assessment, especially when involving dose-sensitive genes such as DMD The pathogenicities of 5'-terminal DMD duplications in the database remain controversial. Four prenatal cases with Xp21.1 duplications were identified by routine prenatal genomic testing, encompassing the 5'-UTR to exons 1-2 in family 1 and family 2, and to exons 1-9 in family 3. The duplication in family 4 was non-contiguous covering the 5'-UTR to exon 1 and exons 3-7. All were traced to unaffected males in the family pedigrees. A new genome-wide approach of optical genome mapping was performed in families 1, 2, and 3 to delineate the breakpoints and orientation of the duplicated fragments. The extra copies were tandemly inserted into the upstream of DMD, preserving the integrity of ORF from the second copy. The pathogenicities were thus reclassified as likely benign. Our data highlight the importance of structural delineation by optical genome mapping in prenatal risk assessment of incidentally identified SVs involving DMD and other similar large dose-sensitive genes.


Sujet(s)
Dystrophine , Pedigree , Humains , Femelle , Mâle , Appréciation des risques/méthodes , Grossesse , Dystrophine/génétique , Diagnostic prénatal/méthodes , Cartographie chromosomique/méthodes , Chromosomes X humains/génétique , Myopathie de Duchenne/génétique , Myopathie de Duchenne/diagnostic , Duplication chromosomique/génétique , Exons/génétique , Duplication de gène/génétique , Adulte
4.
Orphanet J Rare Dis ; 19(1): 311, 2024 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-39182149

RÉSUMÉ

BACKGROUND: An increasing number of clinical trials for new therapeutic strategies are underway or being considered for dystrophinopathy. Having detailed data on the natural progression of this condition is crucial for assessing the effectiveness of new drugs. However, there's a lack of data regarding the long-term data on the natural course and how it's managed in China. In this study, we offer a comprehensive overview of clinical and molecular findings, as well as treatment outcomes in the Chinese population. METHODS: Institutional data on all patients with dystrophinopathy from August 2011 to August 2021 were retrospectively reviewed. The data included geographic distribution, age at diagnosis, molecular findings, and treatment options, such as corticosteroids, cardiac interventions, and clinical outcomes. RESULTS: In total, 2097 patients with dystrophinopathy, including 1703 cases of Duchenne muscular dystrophy (DMD), 311 cases of Becker muscular dystrophy (BMD), 46 cases of intermediate muscular dystrophy (IMD), and 37 cases categorized as "pending" (individuals with an undetermined phenotype), were registered in the Children's Hospital of Fudan University database for dystrophinopathy from August 2011 to August 2021. The spectrum of identified variants included exonic deletions (66.6%), exonic duplications (10.7%), nonsense variants (10.3%), splice-site variants (4.5%), small deletions (3.5%), small insertions/duplications (1.8%), and missense variants (0.9%). Four deep intronic variants and two inversion variants were identified. Regarding treatment, glucocorticoids were administered to 54.4% of DMD patients and 39.1% of IMD patients. The median age at loss of ambulation was 2.5 years later in DMD patients who received glucocorticoid treatment. Overall, one cardiac medicine at least was prescribed to 7.4% of DMD patients, 8.3% of IMD patients, and 2.6% of BMD patients. Additionally, ventilator support was required by four DMD patients. Eligibility for exon skipping therapy was found in 55.3% of DMD patients, with 12.9%, 10%, and 9.6% of these patients being eligible for skipping exons 51, 53, and 45, respectively. CONCLUSIONS: This is one of the largest studies to have evaluated the natural history of dystrophinopathy in China, which is particularly conducive to the recruitment of eligible patients for clinical trials and the provision of real-world data to support drug development.


Sujet(s)
Myopathie de Duchenne , Humains , Myopathie de Duchenne/génétique , Myopathie de Duchenne/traitement médicamenteux , Mâle , Études rétrospectives , Enfant , Femelle , Enfant d'âge préscolaire , Adolescent , Dystrophine/génétique , Chine , Bases de données factuelles , Nourrisson , Dystrophies musculaires/génétique , Dystrophies musculaires/traitement médicamenteux , Jeune adulte
6.
Orphanet J Rare Dis ; 19(1): 260, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982500

RÉSUMÉ

PURPOSE: An increasing number of patients with Duchenne muscular dystrophy (DMD) now have access to improved standard of care and disease modifying treatments, which improve the clinical course of DMD and extend life expectancy beyond 30 years of age. A key issue for adolescent DMD patients is the transition from paediatric- to adult-oriented healthcare. Adolescents and adults with DMD have unique but highly complex healthcare needs associated with long-term steroid use, orthopaedic, respiratory, cardiac, psychological, and gastrointestinal problems meaning that a comprehensive transition process is required. A sub-optimal transition into adult care can have disruptive and deleterious consequences for a patient's long-term care. This paper details the results of a consensus amongst clinicians on transitioning adolescent DMD patients from paediatric to adult neurologists that can act as a guide to best practice to ensure patients have continuous comprehensive care at every stage of their journey. METHODS: The consensus was derived using the Delphi methodology. Fifty-three statements were developed by a Steering Group (the authors of this paper) covering seven topics: Define the goals of transition, Preparing the patient, carers/parents and the adult centre, The transition process at the paediatric centre, The multidisciplinary transition summary - Principles, The multidisciplinary transition summary - Content, First visit in the adult centre, Evaluation of transition. The statements were shared with paediatric and adult neurologists across Central Eastern Europe (CEE) as a survey requesting their level of agreement with each statement. RESULTS: Data from 60 responders (54 full responses and six partial responses) were included in the data set analysis. A consensus was agreed across 100% of the statements. CONCLUSIONS: It is hoped that the findings of this survey which sets out agreed best practice statements, and the transfer template documents developed, will be widely used and so facilitate an effective transition from paediatric to adult care for adolescents with DMD.


Sujet(s)
Méthode Delphi , Myopathie de Duchenne , Humains , Myopathie de Duchenne/thérapie , Adolescent , Israël , Neurologues , Grèce , Adulte , Transition aux soins pour adultes , Consensus , Mâle , Enfant , Femelle , Europe
7.
PLoS One ; 19(7): e0304984, 2024.
Article de Anglais | MEDLINE | ID: mdl-38985784

RÉSUMÉ

Evaluations of treatment efficacy in Duchenne muscular dystrophy (DMD), a rare genetic disease that results in progressive muscle wasting, require an understanding of the 'meaningfulness' of changes in functional measures. We estimated the minimal detectable change (MDC) for selected motor function measures in ambulatory DMD, i.e., the minimal degree of measured change needed to be confident that true underlying change has occurred rather than transient variation or measurement error. MDC estimates were compared across multiple data sources, representing >1000 DMD patients in clinical trials and real-world clinical practice settings. Included patients were ambulatory, aged ≥4 to <18 years and receiving steroids. Minimal clinically important differences (MCIDs) for worsening were also estimated. Estimated MDC thresholds for >80% confidence in true change were 2.8 units for the North Star Ambulatory Assessment (NSAA) total score, 1.3 seconds for the 4-stair climb (4SC) completion time, 0.36 stairs/second for 4SC velocity and 36.3 meters for the 6-minute walk distance (6MWD). MDC estimates were similar across clinical trial and real-world data sources, and tended to be slightly larger than MCIDs for these measures. The identified thresholds can be used to inform endpoint definitions, or as benchmarks for monitoring individual changes in motor function in ambulatory DMD.


Sujet(s)
Myopathie de Duchenne , Myopathie de Duchenne/physiopathologie , Humains , Enfant , Adolescent , Mâle , Enfant d'âge préscolaire , Test de marche , Différence minimale cliniquement importante , Femelle , Marche à pied/physiologie , Activité motrice/physiologie
8.
Article de Anglais | MEDLINE | ID: mdl-38996213

RÉSUMÉ

Duchenne muscular dystrophy (DMD), a genetic condition marked by progressive muscle degeneration, presents notable orthopaedic challenges, especially scoliosis, which deteriorates patients' quality of life by affecting sitting balance and complicating cardiac and respiratory functions. Current orthopaedic management strategies emphasize early intervention with corticosteroids to delay disease progression and the use of surgical spinal fusion to address severe scoliosis, aiming to enhance sitting balance, alleviate discomfort, and potentially extend patient lifespan. Despite advancements, optimal management requires ongoing research to refine therapeutic approaches, ensuring improved outcomes for patients with DMD. This review synthesizes recent findings on surgical and nonsurgical interventions, underscoring the importance of a multidisciplinary approach tailored to the dynamic needs of patients with DMD.


Sujet(s)
Myopathie de Duchenne , Scoliose , Myopathie de Duchenne/chirurgie , Myopathie de Duchenne/thérapie , Humains , Scoliose/chirurgie , Scoliose/thérapie , Arthrodèse vertébrale/méthodes , Qualité de vie , Hormones corticosurrénaliennes/usage thérapeutique , Procédures orthopédiques/méthodes
10.
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
11.
Drugs ; 84(7): 849-856, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38967716

RÉSUMÉ

Givinostat (DUVYZAT™), an orally available histone deacetylase inhibitor, is being developed by Italfarmaco for the treatment of muscular dystrophy and polycythemia vera. Givinostat received its first approval on 21 March 2024, in the USA, for the treatment of Duchenne muscular dystrophy (DMD) in patients 6 years of age and older. Approval was based on the results of the multinational phase III EPIDYS trial, in which givinostat recipients showed less decline than placebo recipients in the time taken to perform a functional task. Givinostat represents the first nonsteroidal treatment for DMD to be approved for use in patients irrespective of the specific genetic variant underlying their disease. Givinostat is available as an oral suspension to be administered twice daily with food. The recommended dosage is based on the body weight of the patient. In the EU, regulatory review of givinostat in DMD is currently underway. This article summarizes the milestones in the development of givinostat leading to this first approval for DMD.


Sujet(s)
Carbamates , Agrément de médicaments , Inhibiteurs de désacétylase d'histone , Myopathie de Duchenne , Humains , Carbamates/administration et posologie , Carbamates/usage thérapeutique , Carbamates/pharmacologie , Myopathie de Duchenne/traitement médicamenteux , Inhibiteurs de désacétylase d'histone/pharmacologie , Inhibiteurs de désacétylase d'histone/administration et posologie , Inhibiteurs de désacétylase d'histone/usage thérapeutique , Inhibiteurs de désacétylase d'histone/effets indésirables , Enfant , États-Unis , Administration par voie orale
12.
Am J Manag Care ; 30(7): e217-e222, 2024 07 01.
Article de Anglais | MEDLINE | ID: mdl-38995826

RÉSUMÉ

OBJECTIVES: To quantify the magnitude of an ISPOR novel value element, insurance value, as applied to new treatments for a rare, severe disease with pediatric onset: Duchenne muscular dystrophy (DMD). STUDY DESIGN: Prospective survey of individuals planning to have children in the future. METHODS: A survey was administered to US adults (aged ≥ 21 years) planning to have a child in the future to elicit willingness to pay (WTP) for insurance coverage for a new hypothetical DMD treatment that improved mortality and morbidity relative to the current standard of care. To identify an indifference point between status quo insurance and insurance with additional cost that would cover the treatment if respondents had a child with DMD, a multiple random staircase design was used. Insurance value-the value individuals receive from a reduction in future health risks-was calculated as the difference between respondent's WTP and what a risk-neutral individual would pay. The risk-neutral value was the product of the (1) probability of having a child with DMD (decision weighted), (2) quality-adjusted life-years (QALYs) gained from the new treatment, and (3) WTP per QALY. RESULTS: Among 207 respondents, 80.2% (n = 166) were aged 25 to 44 years, and 59.9% (n = 124) were women. WTP for insurance coverage of the hypothetical treatment was $973 annually, whereas the decision-weighted risk-neutral value was $452 per year. Thus, insurance value constituted 53.5% ($520) of value for new DMD treatments. CONCLUSIONS: Individuals planning to have children in the future are willing to pay more for insurance coverage of novel DMD treatments than is assumed under risk-neutral, QALY-based frameworks.


Sujet(s)
Myopathie de Duchenne , Maladies rares , Humains , Myopathie de Duchenne/économie , Myopathie de Duchenne/thérapie , Maladies rares/économie , Maladies rares/thérapie , Adulte , Études prospectives , États-Unis , Mâle , Femelle , Assurance maladie/économie , Assurance maladie/statistiques et données numériques , Couverture d'assurance/statistiques et données numériques , Couverture d'assurance/économie , Jeune adulte , Années de vie ajustées sur la qualité , Enfant , Assurance basée sur la valeur/économie
14.
Adv Ther ; 41(8): 3278-3298, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38958840

RÉSUMÉ

INTRODUCTION: Duchenne muscular dystrophy (DMD) is characterized by rapid functional decline. Current available treatment options aim to delay disease progression or stabilize physical function. To aid in healthcare providers' understanding of the symptoms of disease that impact patients' experience, this study explored children's physical functioning, activities of daily living (ADLs), and health-related quality of life (HRQoL) after receiving eteplirsen, a weekly infusion indicated for individuals with DMD with exon 51 skip-amenable mutations. METHODS: Fifteen caregivers of male individuals with DMD participated in a 60-min, semi-structured interview. Open-ended questioning explored changes in the children's condition or maintenance in abilities since eteplirsen initiation. RESULTS: Children with DMD (age 7-15 years [mean 10.9]; steroid treatment at interview, n = 8; time since eteplirsen initiation 3-24 months [mean 14.9]) were described by caregivers as ambulatory (n = 9) and non-ambulatory (n = 6). Caregivers of ambulatory children reported improvements or maintenance of walking ability (n = 7/9), running (n = 6/9), and using stairs (n = 4/9). Continued decline in using stairs was reported by two caregivers. In upper-limb functioning, improvements or maintenances in fine-motor movements were reported by nearly half of all caregivers (n = 7/15), with one caregiver noting a continued decline. Subsequent improvements or maintenances in ADLs were described. Improvements or maintenances in fatigue (n = 9/15), muscle weakness (n = 7/15), and pain (n = 6/15) were reported, although some caregivers described a continued decline (n = 3/15 fatigue, n = 1/15 muscle weakness, n = 2/15 pain). Importantly, most caregivers who reported maintenances in ability perceived this as a positive outcome (n = 6/9). CONCLUSION: This exploratory study indicated that most caregivers perceived improvements or maintenances in aspects of their child's physical functioning, ADLs, and HRQoL since eteplirsen initiation, which they perceived to be a positive outcome.


Duchenne muscular dystrophy (DMD) is a rare disease characterized by progressive muscle weakness. Early on, this weakness presents as difficulty walking, but eventually children lose the ability to walk, develop spinal curvature, and experience problems with the heart and lung muscles. People with DMD are missing a key protein in their bodies called dystrophin. Eteplirsen is a weekly, intravenous treatment approved to treat people with a specific DMD genetic misspelling. The goal of the treatment is to slow down the disease and delay the time to losing ability to walk or needing help breathing. Fifteen caregivers of children living with DMD participated in a 60-min telephone interview. Caregivers were asked questions about the child's DMD symptoms and how those symptoms impact the child's daily life. Caregivers discussed their child's experience while receiving eteplirsen treatment and changes since the start of treatment. Caregivers described their child's muscle weakness and how this has affected their movements (e.g., using stairs, running or walking). Since starting eteplirsen treatment, all caregivers reported some improvement or maintenance in parts of their child's physical functioning, activities of daily living (e.g., sports/leisure, getting dressed and self-care), and symptoms (e.g., muscle weakness, pain and fatigue), even though some decline was also reported (e.g., physical functioning, getting dressed, self-care, muscle weakness, pain and fatigue). The results provide insights into physical functioning and quality of life of children with DMD who are receiving eteplirsen. However, more research is needed to fully understand the impact of eteplirsen on these experiences.


Sujet(s)
Activités de la vie quotidienne , Myopathie de Duchenne , Qualité de vie , Humains , Myopathie de Duchenne/traitement médicamenteux , Myopathie de Duchenne/psychologie , Enfant , Mâle , Adolescent , Aidants/psychologie , Recherche qualitative , Morpholinos
15.
Nutrients ; 16(13)2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38999890

RÉSUMÉ

The progression of Duchenne muscular dystrophy (DMD)requires the assessment of nutritional disturbances at each stage of the disease. The purpose of this study was to assess the nutritional status in various ages of boys with DMD using screening and in-depth evaluation methods. Body composition by Dual X-ray Absorptiometry (DXA), basal metabolic rate (BMR) by indirect calorimetry, a questionnaire of nutritional status-Pediatric Nutrition Screening Tool (PNST)-and laboratory parameters were performed. In the cohort of 93 boys aged 8.54 (5.9-12.6 years), inappropriate nutritional status occurred in 41.8% of boys (underweight 11.8%, overweight 16.0%, and obesity 14.0%). In the 10-13 age group, the occurrence of overweight and underweight was the highest. Based on PNST, 15.1% of patients were at nutritional risk (≥2 points)-the most in the 14-17 age group (29%). A negative correlation was identified between PNST and z-scores of body weight, BMI, and FFMI (r Spearman = -0.49, -0.46, and -0.48, respectively; p < 0.05). There were no differences between BMR results from indirect calorimetry and calculations from the Schofield formula for any age group. In obese boys, the caloric requirement in indirect calorimetry was significantly lower than that indicated by the calculations according to the Schofield formula (p < 0.028). Inappropriate nutritional status occurred in almost half of the children with DMD. The age group in which nutritional disorders were most frequently identified was 10-13 years old. PNST could be considered a tool for screening malnutrition after testing a larger group of DMD patients.


Sujet(s)
Indice de masse corporelle , Myopathie de Duchenne , État nutritionnel , Humains , Myopathie de Duchenne/complications , Myopathie de Duchenne/épidémiologie , Mâle , Enfant , Adolescent , Enfant d'âge préscolaire , Composition corporelle , Évaluation de l'état nutritionnel , Incidence , Maigreur/épidémiologie , Surpoids/épidémiologie , Surpoids/complications , Métabolisme basal , Absorptiométrie photonique , Calorimétrie indirecte , Malnutrition/épidémiologie
16.
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
17.
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
18.
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 , Myopathie de Duchenne , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Animaux , Mâle , Souris , Délétion de gène , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Souris de lignée C57BL , Souris de lignée mdx , Souris knockout , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/génétique , Myopathie de Duchenne/anatomopathologie , Facteur-2 apparenté à NF-E2/métabolisme , Facteur-2 apparenté à NF-E2/génétique , Cellules satellites du muscle squelettique/métabolisme
19.
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
20.
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
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE