1
|
Armytasari I, Sutomo R, Triono A. Longitudinal management in Duchenne muscular dystrophy with exon 63 duplication. BMJ Case Rep 2024; 17:e260706. [PMID: 39353675 DOI: 10.1136/bcr-2024-260706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024] Open
Abstract
A boy with nonambulatory Duchenne muscular dystrophy (DMD) tested positive for exon 63 duplication and exhibited intellectual disability, overweight and dyslipidaemia. The patient underwent a comprehensive multidisciplinary approach involving pharmacological and non-pharmacological interventions. Despite challenges, such as socioeconomic constraints and limited access to advanced therapies, the patient received tailored care. The management included prednisone medication, dietary modifications and psychological support. The patient's journey highlighted the complex interplay of medical and psychosocial factors affecting DMD patients in resource-limited settings. Regular monitoring and the involvement of the patient's family in a peer group were arranged to improve overall quality of life. The case underscores the need for accessible and holistic care for DMD patients, addressing both medical and psychosocial challenges.
Collapse
Affiliation(s)
- Inggar Armytasari
- Department of Child Health, Public Hospital Dr Sardjito, Sleman, Indonesia
- Department of Child Health, Gadjah Mada University, Faculty of Medicine Public Health and Nursing, Sleman, Indonesia
| | - Retno Sutomo
- Department of Child Health, Public Hospital Dr Sardjito, Sleman, Indonesia
- Department of Child Health, Gadjah Mada University, Faculty of Medicine Public Health and Nursing, Sleman, Indonesia
| | - Agung Triono
- Department of Child Health, Public Hospital Dr Sardjito, Sleman, Indonesia
- Department of Child Health, Gadjah Mada University, Faculty of Medicine Public Health and Nursing, Sleman, Indonesia
| |
Collapse
|
2
|
Dhoke NR, Kim H, Azzag K, Crist SB, Kiley J, Perlingeiro RCR. A Novel CRISPR-Cas9 Strategy to Target DYSTROPHIN Mutations Downstream of Exon 44 in Patient-Specific DMD iPSCs. Cells 2024; 13:972. [PMID: 38891104 PMCID: PMC11171783 DOI: 10.3390/cells13110972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Neha R. Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Sarah B. Crist
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Rita C. R. Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
3
|
Pickart AM, Martin AS, Gross BN, Dellefave-Castillo LM, McCallen LM, Nagaraj CB, Rippert AL, Schultz CP, Ulm EA, Armstrong N. Genetic counseling for the dystrophinopathies-Practice resource of the National Society of Genetic Counselors. J Genet Couns 2024. [PMID: 38682751 DOI: 10.1002/jgc4.1892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 03/06/2024] [Accepted: 03/12/2024] [Indexed: 05/01/2024]
Abstract
The dystrophinopathies encompass the phenotypically variable forms of muscular dystrophy caused by pathogenic variants in the DMD gene. The dystrophinopathies include the most common inherited muscular dystrophy among 46,XY individuals, Duchenne muscular dystrophy, as well as Becker muscular dystrophy and other less common phenotypic variants. With increased access to and utilization of genetic testing in the diagnostic and carrier setting, genetic counselors and clinicians in diverse specialty areas may care for individuals with and carriers of dystrophinopathy. This practice resource was developed as a tool for genetic counselors and other health care professionals to support counseling regarding dystrophinopathies, including diagnosis, health risks and management, psychosocial needs, reproductive options, clinical trials, and treatment. Genetic testing efforts have enabled genotype/phenotype correlation in the dystrophinopathies, but have also revealed unexpected findings, further complicating genetic counseling for this group of conditions. Additionally, the therapeutic landscape for dystrophinopathies has dramatically changed with several FDA-approved therapeutics, an expansive research pathway, and numerous clinical trials. Genotype-phenotype correlations are especially complex and genetic counselors' unique skill sets are useful in exploring and explaining this to families. Given the recent advances in diagnostic testing and therapeutics related to dystrophinopathies, this practice resource is a timely update for genetic counselors and other healthcare professionals involved in the diagnosis and care of individuals with dystrophinopathies.
Collapse
Affiliation(s)
- Angela M Pickart
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, District of Columbia, USA
| | - Brianna N Gross
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lisa M Dellefave-Castillo
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Leslie M McCallen
- Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Chinmayee B Nagaraj
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Alyssa L Rippert
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Elizabeth A Ulm
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Niki Armstrong
- Parent Project Muscular Dystrophy, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Gatto F, Benemei S, Piluso G, Bello L. The complex landscape of DMD mutations: moving towards personalized medicine. Front Genet 2024; 15:1360224. [PMID: 38596212 PMCID: PMC11002111 DOI: 10.3389/fgene.2024.1360224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration, with respiratory and cardiac complications, caused by mutations in the DMD gene, encoding the protein dystrophin. Various DMD mutations result in different phenotypes and disease severity. Understanding genotype/phenotype correlations is essential to optimize clinical care, as mutation-specific therapies and innovative therapeutic approaches are becoming available. Disease modifier genes, trans-active variants influencing disease severity and phenotypic expressivity, may modulate the response to therapy, and become new therapeutic targets. Uncovering more disease modifier genes via extensive genomic mapping studies offers the potential to fine-tune prognostic assessments for individuals with DMD. This review provides insights into genotype/phenotype correlations and the influence of modifier genes in DMD.
Collapse
Affiliation(s)
| | | | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| |
Collapse
|
5
|
Savas-Kalender D, Kurt-Aydin M, Acarol FO, Tarsuslu T, Yis U. Dual task impact on functional mobility and interaction of functional level and balance in patients with Duchenne muscular dystrophy. Gait Posture 2024; 108:282-288. [PMID: 38171184 DOI: 10.1016/j.gaitpost.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Dystrophin, a protein crucial for various brain regions governing higher-order functions like learning and memory is notably absent in individuals with Duchenne muscular dystrophy (DMD). This absence of dystrophin in the brain is believed to underlie cognitive challenges in DMD. Cognitive and motor challenges observed in DMD could potentially hinder the execution of dual tasks. RESEARCH QUESTION Is there a significant correlation between dual-task performance, functional mobility, and balance in children with DMD? METHOD The study included 28 participants (14 DMD, 14 typical development). Timed Up and Go (TUG) test results were recorded for single and dual-task conditions (motor-motor, cognitive-motor). Functional level was assessed using Motor Function Measurement-32 (MFM-32), Brooke Upper Extremity Scale, and Vignos Scale. Balance was evaluated using Balance Master System and Pediatric Functional Reach Test (PFRT). RESULTS Significant differences in TUG test scores across conditions were observed in both DMD and typical development groups (p < 0.05). Children with DMD exhibited longer completion times compared to typical development children (p < 0.05). Among children with DMD, there was a significant correlation between TUG scores in different task conditions and balance assessment (p < 0.05, r = 0.571 to -0.819). Lower MFM-32 scores in DMD children were correlated with worse TUG performance across conditions (p < 0.05, r = 0.586 to -0.868). SIGNIFIANCE This study sheds light on the multifaceted nature of dual-tasking challenges in individuals with DMD, thereby contributing to a deeper understanding of the implications for rehabilitation strategies.
Collapse
Affiliation(s)
| | - Merve Kurt-Aydin
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Izmir Katip Celebi University, Izmir, Turkiye
| | | | - Tulay Tarsuslu
- Faculty of Physical Therapy and Rehabilitation, Dokuz Eylul University, Izmir, Turkiye
| | - Uluc Yis
- Department of Pediatric Neurology, Faculty of Medicine, Dokuz Eylül University, İzmir, Turkiye
| |
Collapse
|
6
|
Nakamura A, Matsumura T, Ogata K, Mori‐Yoshimura M, Takeshita E, Kimura K, Kawashima T, Tomo Y, Arahata H, Miyazaki D, Takeshima Y, Takahashi T, Ishigaki K, Kuru S, Wakisaka A, Awano H, Funato M, Sato T, Saito Y, Takada H, Sugie K, Kobayashi M, Ozasa S, Fujii T, Maegaki Y, Oi H, Tachimori H, Komaki H. Natural history of Becker muscular dystrophy: a multicenter study of 225 patients. Ann Clin Transl Neurol 2023; 10:2360-2372. [PMID: 37882106 PMCID: PMC10723226 DOI: 10.1002/acn3.51925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVE Becker muscular dystrophy (BMD) is a milder variant of Duchenne muscular dystrophy (DMD), a lethal X-linked muscular disorder. Here, we aim to investigat the clinical involvement of skeletal, respiratory, cardiac, and central nervous systems in patients with BMD, as well as genotype-phenotype relationships. METHODS This nationwide cohort study investigated the clinical manifestations and genotype-phenotype relationships in 225 patients with BMD having in-frame deletion from 22 medical centers. The primary outcome was to elucidate the association of genotype with skeletal muscle, respiratory, cardiac, and central nervous system disorders. Descriptive statistics were used to analyze the data. RESULTS The average age of the subjects was 31.5 (range, 1-81) years. Initial symptoms of BMD were muscular (60%), followed by asymptomatic hypercreatine kinasemia (32.4%) and central nervous system disorders (5.3%). Gait disturbance was observed in 53.8% of patients and the average age at wheelchair introduction was 36.5 years. The ventilator introduction rate was 6.7% at an average age of 36.6 years. More than 30% of patients had an abnormal electrocardiogram and approximately 15% had heart failure symptoms. Cardiac function on echocardiography varied significantly among the patients. The frequencies of seizures and intellectual/developmental disability were 8.0% and 16.9%, respectively. Exon 45-47deletion (del) was the most common (22.6%), followed by exon 45-48del (13.1%). Patients with exon 45-49del patients demonstrated severe skeletal muscle damage. Patients with exon 45-47del and exon 45-55del patients did not require ventilator use. INTERPRETATION The study provides important prognostic information for patients and clinicians to establish therapy plans and to implement preventative medicine.
Collapse
Affiliation(s)
- Akinori Nakamura
- Department of NeurologyNHO Matsumoto Medical CenterMatsumotoJapan
| | | | - Katsuhisa Ogata
- Department of NeurologyNHO Higashisaitama National HospitalHasudaJapan
| | - Madoka Mori‐Yoshimura
- Department of NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| | - Eri Takeshita
- Department of Child NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| | - Koichi Kimura
- Department of Laboratory Medicine/CardiologyThe Institute of Medical Science, The University of TokyoMinato‐kuJapan
| | - Takahiro Kawashima
- Department of Information Medicine, National Center of Neurology and PsychiatryNational Institute of NeuroscienceKodairaJapan
| | - Yui Tomo
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and PsychiatryKodairaJapan
| | - Hajime Arahata
- Department of Neurology, Neuro‐Muscular CenterNHO Omuta National HospitalOmutaJapan
| | - Daigo Miyazaki
- Department of Medicine (Neurology and Rheumatology)Shinshu University School of MedicineMatsumotoJapan
| | - Yasuhiro Takeshima
- Department of PediatricsHyogo Medical University School of MedicineNishinomiyaJapan
| | | | - Keiko Ishigaki
- Department of PediatricsTokyo Women's Medical University School of MedicineShinjuku‐kuJapan
| | - Satoshi Kuru
- Department of NeurologyNHO Suzuka National HospitalSuzukaJapan
| | - Akiko Wakisaka
- Department of PediatricsNHO Iou National HospitalKanazawaJapan
| | - Hiroyuki Awano
- Research Initiative Center, Organization for Research Initiative and PromotionTottori UniversityYonagoJapan
| | - Michinori Funato
- Department of Pediatric NeurologyNHO Nagara Medical CenterNagaraJapan
| | - Tatsuharu Sato
- Department of PediatricsNagasaki University HospitalNagasakiJapan
| | - Yoshiaki Saito
- Department of PediatricsNational Rehabilitation Center for Children with DisabilitiesItabashiJapan
| | - Hiroto Takada
- Department of NeurologyNHO Aomori National HospitalAomoriJapan
| | - Kazuma Sugie
- Department of NeurologyNara Medical University School of MedicineKashiharaJapan
| | - Michio Kobayashi
- Department of NeurologyNHO Akita National HospitalYurihonjoJapan
| | - Shiro Ozasa
- Department of PediatricsKumamoto University HospitalKumamotoJapan
| | - Tatsuya Fujii
- Department of PediatricsShiga Medical Center for ChildrenMoriyamaJapan
| | - Yoshihiro Maegaki
- Division of Child Neurology, Department of Brain and Neurosciences, Faculty of MedicineTottori UniversityYonagoJapan
| | - Hideki Oi
- Department of Clinical Data Science, Clinical Research & Education Promotion Division, National Center of Neurology and PsychiatryKodairaJapan
| | - Hisateru Tachimori
- Department of Information Medicine, National Center of Neurology and PsychiatryNational Institute of NeuroscienceKodairaJapan
- Endowed Course of Health System InnovationKeio University School of MedicineTokyoJapan
| | - Hirofumi Komaki
- Department of Child NeurologyNational Center Hospital, National Center of Neurology and PsychiatryKodairaJapan
| |
Collapse
|
7
|
Basiri K, Alizadeh M, Ansari B, Ghasemi M, Kheradmand M, Sedghi M. On genotype-phenotype relationship of dystrophinopathies among Iranian population. CURRENT JOURNAL OF NEUROLOGY 2023; 22:231-237. [PMID: 38425356 PMCID: PMC10899535 DOI: 10.18502/cjn.v22i4.14528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/12/2023] [Indexed: 03/02/2024]
Abstract
Background: Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are inherited X-linked disorders resulting from alterations in the dystrophin gene. Genotype-phenotype matching studies have revealed a link between disease severity, the amount of muscle dystrophin, and the extent of mutation/deletion on the dystrophin gene. This study aimed to assess the relationship between genetic alterations in the dystrophin gene and the clinical status of patients with dystrophinopathies among the Iranian population. Methods: This cross-sectional study examined 54 patients with muscle weakness caused by abnormalities in the dystrophin gene at a hospital affiliated to Isfahan University of Medical Sciences, Isfahan, Iran, in 2021. The participants' demographic information, including age, family history of muscle dystrophies, and family history of other medical diseases as well as the type of muscular dystrophy were recorded. Furthermore, the number and region of deleted exons based on dystrophy types were also evaluated using multiplex ligation-dependent probe amplification (MLPA). The patients' gaits were also assessed as using a wheelchair, the presence of waddling gaits, or toe gaits. The patients' clinical status and the coexistence of pulmonary, bulbar, and mental conditions were also examined and compared between the two groups of dystrophinopathies. Results: In this study, 54 patients with dystrophinopathy with the mean age of 16.63 ± 12.10 years were evaluated, of whom 22 (40.7%) and 30 (55.6%) patients were classified as BMD and DMD, respectively. The most affected regions with deleted exons were exons 45-47 (n = 5) and 45-48 (n = 4) in patients with BMD, while exons 45, 48-52, 51-55, and 53 (2 cases per exon) were the most common affected exons in patients with DMD. Further analyses revealed that deletions in exons 45-47 and 51-55 were significantly associated with older and younger ages at the onset of becoming wheelchair-bound in patients with dystrophy, respectively. The hotspot range in both BMD and DMD was within exons 45-55 (n = 15 for each group); 63% of the patients had alterations on the dystrophin gene within this range [30 patients (68.18%) in the BMD group, 15 patients (53.57%) in the DMD group]. Conclusion: Exon deletion was the most common genetic alteration in patients with dystrophinopathies. No significant difference was observed between DMD and BMD regarding the number of deleted exons. Deletions in exons 45-47 and 51-55 were linked to later and earlier onset of becoming wheelchair-bound, respectively.
Collapse
Affiliation(s)
- Keivan Basiri
- Isfahan Neuroscience Research Center, Al-Zahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Alizadeh
- Department of Neurology, School of Medicine, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behnaz Ansari
- Isfahan Neuroscience Research Center, Al-Zahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Majid Ghasemi
- Isfahan Neuroscience Research Center, Al-Zahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohsen Kheradmand
- Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Sedghi
- Medical Genetics Laboratory, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Ishii MN, Quinton M, Kamiguchi H. A highly sensitive and quantitative assay for dystrophin protein using Single Molecule Count Technology. Neuromuscul Disord 2023; 33:737-743. [PMID: 37666691 DOI: 10.1016/j.nmd.2023.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 09/06/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease characterized by progressive muscle loss caused by mutations in dystrophin, resulting in decreased dystrophin levels. Dystrophin protein expression is a biomarker used to evaluate treatments that restore patient dystrophin levels. Currently, a semiquantitative assay using western blotting, which normalizes dystrophin expression to that of a control population, is used for regulatory filing. However, the current methods are limited in terms of sensitivity, quantification, and reproducibility. To address this, a highly sensitive and quantitative sandwich immune assay using Single Molecule Counting technology was established, with recombinant dystrophin protein as the calibrator. Capture and detection antibodies were selected to detect full-length dystrophin. Using this optimized assay, dystrophin levels in muscle samples from Myotonic Dystrophy (n = 9) and DMD (n = 8) subjects were 93.2 ± 31.9 (range: 49.4-145.3) and 14.5 ± 6.8 (range: 6.18-22.6) fmol/total protein mg, respectively. The lowest concentration of dystrophin measured in the DMD samples was 5 times higher than that in the lower limit of quantitation, a level not detected by western blotting. These data indicate that this assay accurately and sensitively measured dystrophin protein and may be useful in clinical trials assessing dystrophin restoration therapies.
Collapse
Affiliation(s)
- Misawa Niki Ishii
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Tokyo, Japan.
| | - Maria Quinton
- Neuroscience Drug Discovery Unit, Takeda Pharmaceuticals, 40 Lansdowne Street, Cambridge MA 02139 USA
| | - Hidenori Kamiguchi
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 251-0012, 26-1, 2-chome, Higashimuraoka, Fujisawa, Kanagawa, Tokyo, Japan
| |
Collapse
|
9
|
Al-Raqad MK, Alwahsh S, Hejazi IS, Abu-Salah OT, Alshadfan L, Abu-Ledeh A, Ghanem N, Braik L, Raggad AD. Electrocardiographic Changes in Jordanian Patients With Becker Muscular Dystrophy. Cureus 2023; 15:e47553. [PMID: 38022137 PMCID: PMC10665640 DOI: 10.7759/cureus.47553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Background and aim Becker muscular dystrophy (BMD) is an X-linked disease caused by an in-frame mutation in the dystrophin gene, which is considered an allelic disorder to the most severe form of dystrophinopahies, Duchenne muscular dystrophy, which leads to skeletal and cardiac muscle involvement and results in dilated cardiomyopathy (DCM). The aim of this study is to present our ECG data and the significance of this data in the early detection of DCM in these patients. Methods This is a retrospective study. All patients known to the clinical Genetic Clinic and Queen Alia Heart Center in Jordan with a diagnosis of Becker muscular dystrophy from the year 2011-2022 are offered cardiac evaluation according to the guidelines, which included clinical assessment, electrocardiograph, and 2-D echocardiograph (echo) at the time of diagnosis and every five years thereafter once the initial assessment was normal. All the records were retrieved and analyzed. Results Fifty-three patients of all ages with genetically confirmed BMD were identified. Twelve had no record as they didn't attend any cardiac evaluation. Forty-one were under regular clinical follow-up. Two were excluded as they died, and another four had no recorded data in our center. Ultimately, 35 patients were included and studied. The mean age was 30.5 years ± 22.1, ranging from two to seventy-seven years of age. Twenty-seven (77%) had abnormal ECG. High voltage R wave in V2 and V1 was the most common finding, followed by repolarisation abnormalities and Q wave (43%, 17%, 13%, and 11% respectively). Incomplete right bundle branch block in 4% as well as R/S ratio >1.2. U wave abnormalities in 3% and sinus tachycardia were found in only one patient. Conclusion Cardiac surveillance for patients with Becker muscular dystrophy is mandatory after the age of 16. Q wave and repolarisation changes should be taken seriously as early signs of dilated cardiomyopathy, even if the echo is normal.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nour Ghanem
- Pediatrics, Al-Balqa Applied University, Al-Salt, JOR
| | - Lana Braik
- Pediatrics, Al-Balqa Applied University, Al-Salt, JOR
| | | |
Collapse
|
10
|
Kavousi S, Pourahmadiyan A, Soleymani F, Noruzinia M. Identification of a Novel de novo Splicing Mutation in Duchenne Muscular Dystrophy Gene in an Iranian Family. Mol Syndromol 2023; 14:331-340. [PMID: 37766830 PMCID: PMC10521217 DOI: 10.1159/000528035] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/05/2022] [Indexed: 09/29/2023] Open
Abstract
Introduction Duchenne muscular dystrophy (DMD) (NM_004006.3) is one of the most notable neuromuscular disorders of early years. The majority of DMD cases are caused by deletions or duplications in dystrophin, while point mutations are less prevalent in dystrophin abnormalities. It is a common knowledge that the severity of the disease depends on the effect of the mutation on the translational reading frame of the dystrophin mRNA. Case Report We studied an 8-year-old boy with relevant clinical presentations for DMD. Deletion/duplication screening was performed by using multiplex ligation-dependent probe amplification, and whole-exome sequencing was conducted in order to identify potential variants. A novel de novo splice site variant was identified in the DMD gene (DMD: c.8548-2A>G). To explore the effect of a novel variant in DMD, various in silico analyses were carried out to investigate the pathogenicity of the causative variant. To study the structure of a DMD protein and information on how the genetic variant impacts splicing site in models of wild-type and mutated DMD, we carried out different computational studies. Sanger sequencing was performed for the purpose of variant confirmation and familial segregation analysis. Discussion This novel de novo variant was predicted to have an effect on splicing, which leads to DMD due to its significant impacts on dystrophin functionality. The novel mutation would be expected to disrupt the protein structure.
Collapse
Affiliation(s)
- Saeideh Kavousi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | - Mehrdad Noruzinia
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
11
|
Ishizuka T, Komaki H, Asahina Y, Nakamura H, Motohashi N, Takeshita E, Shimizu‐Motohashi Y, Ishiyama A, Yonee C, Maruyama S, Hida E, Aoki Y. Systemic administration of the antisense oligonucleotide
NS
‐089/
NCNP
‐02 for skipping of exon 44 in patients with Duchenne muscular dystrophy: Study protocol for a phase I/
II
clinical trial. Neuropsychopharmacol Rep 2023. [DOI: 10.1002/npr2.12335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Affiliation(s)
- Takami Ishizuka
- Clinical Research and Education Promotion Division National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Hirofumi Komaki
- Clinical Research and Education Promotion Division National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
- Department of Child Neurology National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Yasuko Asahina
- Clinical Research and Education Promotion Division National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Harumasa Nakamura
- Clinical Research and Education Promotion Division National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Norio Motohashi
- Department of Molecular Therapy National Institute of Neuroscience, National Center of Neurology and Psychiatry Tokyo Japan
| | - Eri Takeshita
- Department of Child Neurology National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Yuko Shimizu‐Motohashi
- Department of Child Neurology National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Akihiko Ishiyama
- Department of Child Neurology National Center Hospital, National Center of Neurology and Psychiatry Tokyo Japan
| | - Chihiro Yonee
- Department of Pediatrics, Graduate School of Medical and Dental Sciences Kagoshima University Kagoshima City Kagoshima Japan
| | - Shinsuke Maruyama
- Department of Pediatrics, Graduate School of Medical and Dental Sciences Kagoshima University Kagoshima City Kagoshima Japan
| | - Eisuke Hida
- Department of Biostatistics and Data Science, Graduate School of Medicine Osaka University Osaka Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy National Institute of Neuroscience, National Center of Neurology and Psychiatry Tokyo Japan
| |
Collapse
|
12
|
Schreyer L, Reilly J, McConkey H, Kerkhof J, Levy MA, Hu J, Hnaini M, Sadikovic B, Campbell C. The discovery of the DNA methylation episignature for Duchenne muscular dystrophy. Neuromuscul Disord 2023; 33:5-14. [PMID: 36572586 DOI: 10.1016/j.nmd.2022.12.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/09/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is an X-linked recessive neuromuscular disorder characterized by progressive muscle weakness due to loss of function mutations in the dystrophin gene. Variation in clinical presentation, the rate of disease progression, and treatment responsiveness have been observed amongst DMD patients, suggesting that factors beyond the loss of dystrophin may contribute to DMD pathophysiology. Epigenetic mechanisms are becoming recognized as important factors implicated in the etiology and progression of various diseases. A growing number of genetic syndromes have been associated with unique genomic DNA methylation patterns (called "episignatures") that can be used for diagnostic testing and as disease biomarkers. To further investigate DMD pathophysiology, we assessed the genome-wide DNA methylation profiles of peripheral blood from 36 patients with DMD using the combination of Illumina Infinium Methylation EPIC bead chip array and EpiSign technology. We identified a unique episignature for DMD that whose specificity was confirmed in relation other neurodevelopmental disorders with known episignatures. By modeling the DMD episignature, we developed a new DMD episignature biomarker and provided novel insights into the molecular pathogenesis of this disorder, which have the potential to advance more effective, personalized approaches to DMD care.
Collapse
Affiliation(s)
- Leighton Schreyer
- Department of Neuroscience, Western University, London, ON N6A 3K7, Canada
| | - Jack Reilly
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Haley McConkey
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jennifer Kerkhof
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Michael A Levy
- Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada
| | - Jonathan Hu
- Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mona Hnaini
- Department of Pediatrics, Clinical Neurological Sciences, Western University, London, ON N6A 3K7, Canada
| | - Bekim Sadikovic
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 3K7, Canada; Verspeeten Clinical Genome Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada.
| | - Craig Campbell
- Department of Pediatrics, Clinical Neurological Sciences and Epidemiology, Western University, London, ON N6A 3K7, Canada.
| |
Collapse
|
13
|
Kurt M, Savaş D, Şimşek TT, Yiş U. Factors associated with balance ability in Duchenne and Becker muscular dystrophies. Gait Posture 2023; 99:139-145. [PMID: 36435068 DOI: 10.1016/j.gaitpost.2022.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/08/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Problems such as progressive muscle function loss, postural deteriorations, and contractures seen in patients with Duchenne and Becker muscular dystrophies (D/BMD) may affect children's balance ability, and impaired balance ability may lead to limitations in children's functional level. RESEARCH QUESTION What factors are associated with balance ability in children with D/BMD? METHODS Twenty participants with D/BMD were included in the study. Participants' ages were asked; height, body weight, and body mass index (BMI) were recorded. Children's functional level was assessed with the Motor Function Measurement-32 (MFM-32), Brooke and Vignos Scales. Muscle strength of hip flexion and extension, knee flexion and extension, and ankle dorsiflexion was measured with a handheld dynamometer. Balance measurements were performed using the Balance Master System. The relationship between balance and continuous independent variables was determined using Spearman's test. RESULTS The mean age of the participants was 8.57 ± 3.27 years. The balance abilities of children with BMD were better than those of children with DMD (p < 0.05). The balance diminished with age (p < 0.05), while there was a positive correlation between balance and weight, height, BMI (p < 0.05). There was a positive correlation between the balance and the MFM-32 total and subsection scores. Muscle strength was positively related to balance (p < 0.05). SIGNIFICANCE The results showed the balance ability in children with D/BMD was affected by age, height, weight, BMI, functional level, and muscle strength. Based on the results of this study, balance and strength training should be an integral part of the rehabilitation of children with D/BMD.
Collapse
Affiliation(s)
- Merve Kurt
- Department of Physiotherapy and Rehabilitation, Faculty of Health Sciences, Izmir Katip Celebi University, Izmir, Turkey; Graduate School of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Dilan Savaş
- Graduate School of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Tülay Tarsuslu Şimşek
- Faculty of Physical Therapy and Rehabilitation, Dokuz Eylul University, Izmir, Turkey.
| | - Uluç Yiş
- Dokuz Eylül University Department of Pediatrics, Pediatric Neurology, İzmir, Turkey.
| |
Collapse
|
14
|
Gomez Limia C, Baird M, Schwartz M, Saxena S, Meyer K, Wein N. Emerging Perspectives on Gene Therapy Delivery for Neurodegenerative and Neuromuscular Disorders. J Pers Med 2022; 12:1979. [PMID: 36556200 PMCID: PMC9788053 DOI: 10.3390/jpm12121979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD) and Parkinson's Disease (PD), are a group of heterogeneous diseases that mainly affect central nervous system (CNS) functions. A subset of NDDs exhibit CNS dysfunction and muscle degeneration, as observed in Gangliosidosis 1 (GM1) and late stages of PD. Neuromuscular disorders (NMDs) are a group of diseases in which patients show primary progressive muscle weaknesses, including Duchenne Muscular Dystrophy (DMD), Pompe disease, and Spinal Muscular Atrophy (SMA). NDDs and NMDs typically have a genetic component, which affects the physiological functioning of critical cellular processes, leading to pathogenesis. Currently, there is no cure or efficient treatment for most of these diseases. More than 200 clinical trials have been completed or are currently underway in order to establish safety, tolerability, and efficacy of promising gene therapy approaches. Thus, gene therapy-based therapeutics, including viral or non-viral delivery, are very appealing for the treatment of NDDs and NMDs. In particular, adeno-associated viral vectors (AAV) are an attractive option for gene therapy for NDDs and NMDs. However, limitations have been identified after systemic delivery, including the suboptimal capacity of these therapies to traverse the blood-brain barrier (BBB), degradation of the particles during the delivery, high reactivity of the patient's immune system during the treatment, and the potential need for redosing. To circumvent these limitations, several preclinical and clinical studies have suggested intrathecal (IT) delivery to target the CNS and peripheral organs via cerebrospinal fluid (CSF). CSF administration can vastly improve the delivery of small molecules and drugs to the brain and spinal cord as compared to systemic delivery. Here, we review AAV biology and vector design elements, different therapeutic routes of administration, and highlight CSF delivery as an attractive route of administration. We discuss the different aspects of neuromuscular and neurodegenerative diseases, such as pathogenesis, the landscape of mutations, and the biological processes associated with the disease. We also describe the hallmarks of NDDs and NMDs as well as discuss current therapeutic approaches and clinical progress in viral and non-viral gene therapy and enzyme replacement strategies for those diseases.
Collapse
Affiliation(s)
- Cintia Gomez Limia
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Megan Baird
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Maura Schwartz
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Smita Saxena
- Department of Neurology, Inselspital, 3010 Bern, Switzerland
| | - Kathrin Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
15
|
Abstract
Muscular dystrophies are a group of genetic disorders characterized by varying degrees of progressive muscle weakness and degeneration. They are clinically and genetically heterogeneous but share the common histological features of dystrophic muscle. There is currently no cure for muscular dystrophies, which is of particular concern for the more disabling and/or lethal forms of the disease. Through the years, several therapies have encouragingly been developed for muscular dystrophies and include genetic, cellular, and pharmacological approaches. In this chapter, we undertake a comprehensive exploration of muscular dystrophy therapeutics under current development. Our review includes antisense therapy, CRISPR, gene replacement, cell therapy, nonsense suppression, and disease-modifying small molecule compounds.
Collapse
|
16
|
Lambrescu I, Popa A, Manole E, Ceafalan LC, Gaina G. Application of Droplet Digital PCR Technology in Muscular Dystrophies Research. Int J Mol Sci 2022; 23:ijms23094802. [PMID: 35563191 PMCID: PMC9099497 DOI: 10.3390/ijms23094802] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022] Open
Abstract
Although they are considered rare disorders, muscular dystrophies have a strong impact on people’s health. Increased disease severity with age, frequently accompanied by the loss of ability to walk in some people, and the lack of treatment, have directed the researchers towards the development of more effective therapeutic strategies aimed to improve the quality of life and life expectancy, slow down the progression, and delay the onset or convert a severe phenotype into a milder one. Improved understanding of the complex pathology of these diseases together with the tremendous advances in molecular biology technologies has led to personalized therapeutic procedures. Different approaches that are currently under extensive investigation require more efficient, sensitive, and less invasive methods. Due to its remarkable analytical sensitivity, droplet digital PCR has become a promising tool for accurate measurement of biomarkers that monitor disease progression and quantification of various therapeutic efficiency and can be considered a tool for non-invasive prenatal diagnosis and newborn screening. Here, we summarize the recent applications of droplet digital PCR in muscular dystrophy research and discuss the factors that should be considered to get the best performance with this technology.
Collapse
Affiliation(s)
- Ioana Lambrescu
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Alexandra Popa
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Animal Production and Public Health, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania
| | - Emilia Manole
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Laura Cristina Ceafalan
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (I.L.); (A.P.); (E.M.); (L.C.C.)
- Correspondence: ; Tel.: +40-21-319-2732
| |
Collapse
|
17
|
Diversity of Dystrophin Gene Mutations and Disease Progression in a Contemporary Cohort of Duchenne Muscular Dystrophy. Pediatr Cardiol 2022; 43:855-867. [PMID: 35064276 DOI: 10.1007/s00246-021-02797-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 11/24/2021] [Indexed: 01/14/2023]
Abstract
Abnormal dystrophin production due to mutations in the dystrophin gene causes Duchenne Muscular Dystrophy (DMD). Cases demonstrate considerable genetic and disease progression variability. It is unclear if specific gene mutations are prognostic of outcomes in this population. We conducted a retrospective cohort study of DMD patients followed at 17 centers across the USA and Canada from 2005 to 2015 with goal of understanding the genetic variability of DMD and its impact on clinical outcomes. Cumulative incidence of clinically relevant outcomes was stratified by genetic mutation type, exon mutation location, and extent of exon deletion. Of 436 males with DMD, 324 (74.3%) underwent genetic testing. Deletions were the most common mutation type (256, 79%), followed by point mutations (45, 13.9%) and duplications (23, 7.1%). There were 131 combinations of mutations with most mutations located along exons 45 to 52. The number of exons deleted varied between 1 and 52 with a median of 3 exons deleted (IQR 1-6). Subjects with mutations starting at exon positions 40-54 had a later onset of arrhythmias occurring at median age 25 years (95% CI 18-∞), p = 0.01. Loss of ambulation occurred later at median age of 13 years (95% CI 12-15) in subjects with mutations that started between exons 55-79, p = 0.01. There was no association between mutation type or location and onset of cardiac dysfunction. We report the genetic variability in DMD and its association with timing of clinical outcomes. Genetic modifiers may explain some phenotypic variability.
Collapse
|
18
|
Development of DG9 peptide-conjugated single- and multi-exon skipping therapies for the treatment of Duchenne muscular dystrophy. Proc Natl Acad Sci U S A 2022; 119:2112546119. [PMID: 35193974 PMCID: PMC8892351 DOI: 10.1073/pnas.2112546119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2021] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disorder of progressive body-wide muscle weakness, considered the most common muscular dystrophy worldwide. Most patients have out-of-frame deletions in the DMD gene, leading to dystrophin absence in muscle. There is no cure for DMD, but exon skipping is emerging as a potential therapy that uses antisense oligonucleotides to convert out-of-frame to in-frame mutations, enabling the production of truncated, partially functional dystrophin. Currently approved exon skipping therapies, however, have limited applicability and efficacy. Here, we developed a more economical approach to skip DMD exons 45 to 55 (a strategy that could treat nearly half of all DMD patients) and identified DG9 peptide conjugation as a powerful way to improve exon skipping efficiencies in vivo. Duchenne muscular dystrophy (DMD) is primarily caused by out-of-frame deletions in the dystrophin gene. Exon skipping using phosphorodiamidate morpholino oligomers (PMOs) converts out-of-frame to in-frame mutations, producing partially functional dystrophin. Four single-exon skipping PMOs are approved for DMD but treat only 8 to 14% of patients each, and some exhibit poor efficacy. Alternatively, exons 45 to 55 skipping could treat 40 to 47% of all patients and is associated with improved clinical outcomes. Here, we report the development of peptide-conjugated PMOs for exons 45 to 55 skipping. Experiments with immortalized patient myotubes revealed that exons 45 to 55 could be skipped by targeting as few as five exons. We also found that conjugating DG9, a cell-penetrating peptide, to PMOs improved single-exon 51 skipping, dystrophin restoration, and muscle function in hDMDdel52;mdx mice. Local administration of a minimized exons 45 to 55–skipping DG9-PMO mixture restored dystrophin production. This study provides proof of concept toward the development of a more economical and effective exons 45 to 55–skipping DMD therapy.
Collapse
|
19
|
Lim KRQ, Shah MNA, Woo S, Wilton-Clark H, Zhabyeyev P, Wang F, Maruyama R, Oudit GY, Yokota T. Natural History of a Mouse Model Overexpressing the Dp71 Dystrophin Isoform. Int J Mol Sci 2021; 22:ijms222312617. [PMID: 34884423 PMCID: PMC8657860 DOI: 10.3390/ijms222312617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 11/18/2022] Open
Abstract
Dystrophin is a 427 kDa protein that stabilizes muscle cell membranes through interactions with the cytoskeleton and various membrane-associated proteins. Loss of dystrophin as in Duchenne muscular dystrophy (DMD) causes progressive skeletal muscle weakness and cardiac dysfunction. Multiple promoters along the dystrophin gene (DMD) give rise to a number of shorter isoforms. Of interest is Dp71, a 71 kDa isoform implicated in DMD pathology by various animal and patient studies. Strong evidence supporting such a role for Dp71, however, is lacking. Here, we use del52;WT mice to understand how Dp71 overexpression affects skeletal and cardiac muscle phenotypes. Apart from the mouse Dmd gene, del52;WT mice are heterozygous for a full-length, exon 52-deleted human DMD transgene expected to only permit Dp71 expression in muscle. Thus, del52;WT mice overexpress Dp71 through both the human and murine dystrophin genes. We observed elevated Dp71 protein in del52;WT mice, significantly higher than wild-type in the heart but not the tibialis anterior. Moreover, del52;WT mice had generally normal skeletal muscle but impaired cardiac function, exhibiting significant systolic dysfunction as early as 3 months. No histological abnormalities were found in the tibialis anterior and heart. Our results suggest that Dp71 overexpression may have more detrimental effects on the heart than on skeletal muscles, providing insight into the role of Dp71 in DMD pathogenesis.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
| | - Md Nur Ahad Shah
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
| | - Stanley Woo
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
| | - Harry Wilton-Clark
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
| | - Pavel Zhabyeyev
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2G3, Canada; (P.Z.); (F.W.)
| | - Faqi Wang
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2G3, Canada; (P.Z.); (F.W.)
| | - Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
| | - Gavin Y. Oudit
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2G3, Canada; (P.Z.); (F.W.)
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB T6G2B7, Canada
- Correspondence: (G.Y.O.); (T.Y.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (M.N.A.S.); (S.W.); (H.W.-C.); (R.M.)
- Muscular Dystrophy Canada Research Chair, Edmonton, AB T6G2H7, Canada
- Correspondence: (G.Y.O.); (T.Y.)
| |
Collapse
|
20
|
Genomic Aberrations Associated with the Pathophysiological Mechanisms of Neurodevelopmental Disorders. Cells 2021; 10:cells10092317. [PMID: 34571966 PMCID: PMC8470284 DOI: 10.3390/cells10092317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/30/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022] Open
Abstract
Genomic studies are increasingly revealing that neurodevelopmental disorders are caused by underlying genomic alterations. Chromosomal microarray testing has been used to reliably detect minute changes in genomic copy numbers. The genes located in the aberrated regions identified in patients with neurodevelopmental disorders may be associated with the phenotypic features. In such cases, haploinsufficiency is considered to be the mechanism, when the deletion of a gene is related to neurodevelopmental delay. The loss-of-function mutation in such genes may be evaluated using next-generation sequencing. On the other hand, the patients with increased copy numbers of the genes may exhibit different clinical symptoms compared to those with loss-of-function mutation in the genes. In such cases, the additional copies of the genes are considered to have a dominant negative effect, inducing cell stress. In other cases, not the copy number changes, but mutations of the genes are responsible for causing the clinical symptoms. This can be explained by the dominant negative effects of the gene mutations. Currently, the diagnostic yield of genomic alterations using comprehensive analysis is less than 50%, indicating the existence of more subtle alterations or genomic changes in the untranslated regions. Copy-neutral inversions and insertions may be related. Hence, better analytical algorithms specialized for the detection of such alterations are required for higher diagnostic yields.
Collapse
|
21
|
Understanding Neuromuscular Health and Disease: Advances in Genetics, Omics, and Molecular Function. J Pers Med 2021; 11:jpm11050438. [PMID: 34065209 PMCID: PMC8161133 DOI: 10.3390/jpm11050438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 05/18/2021] [Indexed: 11/24/2022] Open
|
22
|
A Genotype-Phenotype Correlation Study of Exon Skip-Equivalent In-Frame Deletions and Exon Skip-Amenable Out-of-Frame Deletions across the DMD Gene to Simulate the Effects of Exon-Skipping Therapies: A Meta-Analysis. J Pers Med 2021; 11:jpm11010046. [PMID: 33466756 PMCID: PMC7830903 DOI: 10.3390/jpm11010046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 01/16/2023] Open
Abstract
Dystrophinopathies are caused by mutations in the DMD gene. Out-of-frame deletions represent most mutational events in severe Duchenne muscular dystrophy (DMD), while in-frame deletions typically lead to milder Becker muscular dystrophy (BMD). Antisense oligonucleotide-mediated exon skipping converts an out-of-frame transcript to an in-frame one, inducing a truncated but partially functional dystrophin protein. The reading frame rule, however, has many exceptions. We thus sought to simulate clinical outcomes of exon-skipping therapies for DMD exons from clinical data of exon skip-equivalent in-frame deletions, in which the expressed quasi-dystrophins are comparable to those resulting from exon-skipping therapies. We identified a total of 1298 unique patients with exon skip-equivalent mutations in patient registries and the existing literature. We classified them into skip-equivalent deletions of each exon and statistically compared the ratio of DMD/BMD and asymptomatic individuals across the DMD gene. Our analysis identified that five exons are associated with significantly milder phenotypes than all other exons when corresponding exon skip-equivalent in-frame deletion mutations occur. Most exon skip-equivalent in-frame deletions were associated with a significantly milder phenotype compared to corresponding exon skip-amenable out-of-frame mutations. This study indicates the importance of genotype-phenotype correlation studies in the rational design of exon-skipping therapies.
Collapse
|