1
|
Gorgoglione D, Sabbatini D, Riguzzi P, Capece G, Pane M, Servidei S, Briganti M, Sancricca C, Bruschi F, Ardissone A, Masson R, Gallone A, Maggi L, Picillo E, Politano L, Petrosino A, Vianello S, Penzo M, Villa M, Sframeli M, Allegra C, Barp A, Di Bari A, Salmin F, Albamonte E, Colacicco G, Panicucci C, Traverso M, Palermo C, Lerario A, Velardo D, D'Angelo MG, Berardinelli A, Gardani A, Nicotra R, Parravicini S, Siciliano G, Ricci G, Torri F, Gadaleta G, Urbano G, Rolle E, Ricci F, D'Amico A, Catteruccia M, Pini A, Giannotta M, Battini R, Marinella G, Previtali SC, Zambon AA, Ferlini A, Fortunato F, Magri F, Mongini TE, Sansone VA, Bruno C, Messina S, Nigro V, Moroni I, Mercuri E, Bello L, Pegoraro E. Natural history of Becker muscular dystrophy: DMD gene mutations predict clinical severity. Brain 2025; 148:1695-1706. [PMID: 39499670 DOI: 10.1093/brain/awae358] [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: 07/08/2024] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Becker muscular dystrophy (BMD) is an X-linked neuromuscular disease attributable to mutations in DMD, leading to a deficient and less functional dystrophin, mainly in skeletal and cardiac muscle. Understanding the natural history of BMD is crucial for optimizing patient care and developing targeted treatments. Retrospective data were collected from 943 patients diagnosed with BMD based on a combination of clinical, biochemical and genetic criteria followed by 17 Italian neuromuscular centres. Patients' demographics, main signs and symptoms at BMD onset, neuropsychiatric comorbidities, age at loss of ambulation, cardiac left ventricular ejection fraction, pulmonary forced vital capacity and DMD mutations were collected. Disease milestones were analysed in specific DMD mutational groups. The median age at the last assessment was 26.0 (16.6-41.9) years, with a median age at diagnosis of 7.5 (4.0-14.0) years. In 55% of patients, the diagnosis was prompted by the incidental finding of hyperCKaemia. At the last assessment, 13.5% of patients had lost the ability to walk at a median age estimated by Kaplan-Meier analysis of 69 years. Thirty per cent of patients exhibited left ventricular impairment and 2.7% respiratory involvement. Ten per cent of patients carried out-of-frame mutations, 4% nonsense mutations and 86% in-frame deletions/duplications. The subset of in-frame deletions was classified further based on the specific mutations. Patients carrying del45-49 compared with del45-47 were associated with an earlier loss of ambulation (P = 1 × 10-4), whereas patients with del45-55 (P = 0.005), del48 (P = 0.02) and del48-49 (P = 0.02) were correlated with a later loss of ambulation compared with del45-47. Both del45-55 (P = 0.002) and del48 (P = 0.003) were significantly associated with decreased odds of developing a pathological left ventricular ejection fraction compared with del45-47. Our results contribute to a better understanding of the natural history of BMD and capture precious data in the era of emerging therapies. The knowledge of the specific DMD mutation might help to define a prognosis in a subset of BMD patients and will serve as a model for the design of future therapies.
Collapse
Affiliation(s)
- Domenico Gorgoglione
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Daniele Sabbatini
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
- Unit of Biostatistics, Epidemiology and Public Health, Department of Cardiac, Thoracic, Vascular Sciences, and Public Health, University of Padova, Padova 35131, Italy
| | - Pietro Riguzzi
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Giuliana Capece
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Roma 00168, Italy
- Centro Clinico Nemo Pediatrico, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma 00168, Italy
| | - Serenella Servidei
- Neurophysiopathology Unit, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
- Dipartimento Universitario di Neuroscienze, Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Marta Briganti
- Neurophysiopathology Unit, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
- Fondazione UILDM Lazio Onlus, Rome 00167, Italy
| | - Cristina Sancricca
- Neurophysiopathology Unit, Dipartimento di Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome 00168, Italy
- Fondazione UILDM Lazio Onlus, Rome 00167, Italy
| | - Fabio Bruschi
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Anna Ardissone
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Riccardo Masson
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Annamaria Gallone
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Esther Picillo
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Napoli 80138, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania 'Luigi Vanvitelli', Napoli 80138, Italy
| | - Angela Petrosino
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Sara Vianello
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Martina Penzo
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Matteo Villa
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Maria Sframeli
- Department of Clinical and Experimental Medicine, University of Messina, Messina 98122, Italy
| | - Cosimo Allegra
- Department of Clinical and Experimental Medicine, University of Messina, Messina 98122, Italy
| | - Andrea Barp
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Alessandra Di Bari
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Francesca Salmin
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Emilio Albamonte
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Giovanni Colacicco
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Chiara Panicucci
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Monica Traverso
- Pediatric Neurology and Muscle Disease Unit, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
| | - Concetta Palermo
- Centro Clinico Nemo Pediatrico, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma 00168, Italy
| | - Alberto Lerario
- Neuromuscular and Rare Disease Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Daniele Velardo
- Neuromuscular and Rare Disease Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Maria G D'Angelo
- Unit of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini 23842, Italy
| | - Angela Berardinelli
- Child and Adolescence Neuromuscular Disorders Research Section, IRCCS Mondino Foundation, Pavia 27100, Italy
| | - Alice Gardani
- Child and Adolescence Neuromuscular Disorders Research Section, IRCCS Mondino Foundation, Pavia 27100, Italy
| | - Roberta Nicotra
- Child and Adolescence Neurological Unit, National Neurological Institute Casimiro Mondino Foundation, IRCCS, IRCCS Mondino Foundation, Pavia 27100, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - Stefano Parravicini
- Child and Adolescence Neurological Unit, National Neurological Institute Casimiro Mondino Foundation, IRCCS, IRCCS Mondino Foundation, Pavia 27100, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Francesca Torri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
| | - Giulio Gadaleta
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Turin 10126, Italy
| | - Guido Urbano
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Turin 10126, Italy
| | - Enrica Rolle
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Turin 10126, Italy
| | - Federica Ricci
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Turin 10126, Italy
| | - Adele D'Amico
- IRCCS, Unit of Muscular and Neurodegenerative disorders, Bambino Gesù Children's Hospital, Rome 00165, Italy
| | - Michela Catteruccia
- IRCCS, Unit of Muscular and Neurodegenerative disorders, Bambino Gesù Children's Hospital, Rome 00165, Italy
| | - Antonella Pini
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Neuromuscular Pediatric Unit, Bologna 40139, Italy
| | - Melania Giannotta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Neuromuscular Pediatric Unit, Bologna 40139, Italy
| | - Roberta Battini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56126, Italy
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa 56128, Italy
| | - Gemma Marinella
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Pisa 56128, Italy
| | - Stefano C Previtali
- Neuromuscular Repair Unit, Division of Neuroscience, Institute of Experimental Neurology (InSpe), IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Alberto A Zambon
- Neuromuscular Repair Unit, Division of Neuroscience, Institute of Experimental Neurology (InSpe), IRCCS Ospedale San Raffaele, Milan 20132, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Fernanda Fortunato
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara 44121, Italy
| | - Francesca Magri
- Neurology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Tiziana E Mongini
- Neuromuscular Unit, Department of Neurosciences RLM, University of Torino, Turin 10126, Italy
| | - Valeria A Sansone
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan 20162, Italy
| | - Claudio Bruno
- Center of Translational and Experimental Myology, IRCCS Istituto Giannina Gaslini, Genova 16147, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health-DINOGMI, University of Genova, Genova 16132, Italy
| | - Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina 98122, Italy
| | - Vincenzo Nigro
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Napoli 80138, Italy
- Telethon Institute of Genetics and Medicine, Pozzuoli 80078, Italy
| | - Isabella Moroni
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico 'Carlo Besta', Milan 20133, Italy
| | - Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Roma 00168, Italy
- Centro Clinico Nemo Pediatrico, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma 00168, Italy
| | - Luca Bello
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| | - Elena Pegoraro
- Department of Neurosciences, Neuromuscular Center, University of Padova, Padova 35128, Italy
| |
Collapse
|
2
|
Sarvutiene J, Ramanavicius A, Ramanavicius S, Prentice U. Advances in Duchenne Muscular Dystrophy: Diagnostic Techniques and Dystrophin Domain Insights. Int J Mol Sci 2025; 26:3579. [PMID: 40332074 PMCID: PMC12027135 DOI: 10.3390/ijms26083579] [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: 02/11/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Abnormalities in X chromosomes, either numerical or structural, cause X-linked disorders, such as Duchenne muscular dystrophy (DMD). Recent molecular and cytogenetic techniques can help identify DMD gene mutations. The accurate diagnosis of Duchenne is crucial, directly impacting patient treatment management, genetics, and the establishment of effective prevention strategies. This review provides an overview of X chromosomal disorders affecting Duchenne and discusses how mutations in Dystrophin domains can impact detection accuracy. Firstly, the efficiency and use of cytogenetic and molecular techniques for the genetic diagnosis of Duchenne disease have, thus, become increasingly important. Secondly, artificial intelligence (AI) will be instrumental in developing future therapies by enabling the aggregation and synthesis of extensive and heterogeneous datasets, thereby elucidating underlying molecular mechanisms. However, despite advances in diagnostic technology, understanding the role of Dystrophin in Duchenne disease remains a challenge. Therefore, this review aims to synthesize this complex information to significantly advance the understanding of DMD and how it could affect patient care.
Collapse
Affiliation(s)
- Julija Sarvutiene
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
| | - Arunas Ramanavicius
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko St. 24, LT-03225 Vilnius, Lithuania
| | - Simonas Ramanavicius
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
| | - Urte Prentice
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko St. 24, LT-03225 Vilnius, Lithuania
- Department of Personalised Medicine, State Research Institute Center for Innovative Medicine, Santariskiu St. 5, LT-08410 Vilnius, Lithuania
| |
Collapse
|
3
|
Tang A, Yokota T. Is Duchenne gene therapy a suitable treatment despite its immunogenic class effect? Expert Opin Drug Saf 2025; 24:395-411. [PMID: 39720847 DOI: 10.1080/14740338.2024.2447072] [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: 05/21/2024] [Revised: 11/22/2024] [Accepted: 12/22/2024] [Indexed: 12/26/2024]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a severe X-linked disorder characterized by progressive muscle weakness and eventual death due to cardiomyopathy or respiratory complications. Currently, there is no cure for DMD, with standard treatments primarily focusing on symptom management. Using immunosuppressive measures and optimized vector designs allows for gene therapies to better address the genetic cause of the disease. AREAS COVERED This review evaluates the efficacy and safety of emerging DMD gene therapies as of 2024. It also discusses the potential of utrophin upregulation, gene editing, and truncated dystrophin as therapeutic strategies. It highlights safety concerns associated with these therapies, including adverse events and patient deaths. A comprehensive overview of developments covers topics such as CRISPR-Cas9 therapies, micro-dystrophin, and the potential delivery of full-length dystrophin. EXPERT OPINION The FDA's recent approval of delandistrogene moxeparvovec (Elevidys) underscores the promise of gene replacement therapies for DMD patients. Understanding the mechanisms behind the adverse effects and excluding patients with specific pathogenic variants may enhance the safety profiles of these therapies. CRISPR/Cas9 therapies, while promising, face significant regulatory and safety challenges that hinder their clinical application. Optimal DMD therapies should target both skeletal and cardiac muscles to be effective.
Collapse
Affiliation(s)
- Annie Tang
- Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Toshifumi Yokota
- Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Miyazaki D, Sato M, Shiba N, Yoshizawa T, Nakamura A. Becker muscular dystrophy mice showed site-specific decay of type IIa fibers with capillary change in skeletal muscle. eLife 2025; 13:RP100665. [PMID: 40094282 PMCID: PMC11913446 DOI: 10.7554/elife.100665] [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] [Indexed: 03/19/2025] Open
Abstract
Becker muscular dystrophy (BMD), an X-linked muscular dystrophy, is mostly caused by an in-frame deletion of Duchenne muscular dystrophy (DMD). BMD severity varies from asymptomatic to severe, associated with the genotype of DMD. However, the underlying mechanisms remain unclear. We established BMD mice carrying three representative exon deletions: ex45-48 del., ex45-47 del., and ex45-49 del. (d45-48, d45-47, and d45-49), with high frequencies and different severities in the human BMD hotspot. All three BMD mice showed muscle weakness, muscle degeneration, and fibrosis, but these changes appeared at different times for each exon deletion, consistent with the severities obtained by the natural history study of BMD. BMD mice showed site-specific muscle changes, unlike mdx mice, which showed diffuse muscle changes, and we demonstrated selective type IIa fiber reduction in BMD mice. Furthermore, BMD mice showed sarcolemmal neuronal nitric oxide synthase (nNOS) reduction and morphological capillary changes around type IIa fibers. These results suggest that capillary changes caused by nNOS reduction may be associated with the mechanism of skeletal muscle degeneration and type IIa fiber reduction in BMD mice. BMD mice may be useful in elucidating the pathomechanisms and developing vascular targeted therapies for human BMD.
Collapse
Affiliation(s)
- Daigo Miyazaki
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of MedicineMatsumotoJapan
- Intractable Disease Care Center, Shinshu University HospitalMatsumotoJapan
| | - Mitsuto Sato
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of MedicineMatsumotoJapan
- Department of Brain Disease Research, Shinshu University School of MedicineMatsumotoJapan
| | - Naoko Shiba
- Department of Regenerative Science and Medicine, Shinshu UniversityMatsumotoJapan
| | - Takahiro Yoshizawa
- Research Center for Advanced Science and Technology, Shinshu UniversityMatsumotoJapan
| | - Akinori Nakamura
- Department of Clinical Research, NHO Matsumoto Medical CenterMatsumotoJapan
- Third Department of Medicine, Shinshu University School of MedicineMatsumotoJapan
| |
Collapse
|
5
|
Stemmerik MG, Tasca G, Gilhus NE, Servais L, Vicino A, Maggi L, Sansone V, Vissing J. Biological biomarkers in muscle diseases relevant for follow-up and evaluation of treatment. Brain 2025; 148:363-375. [PMID: 39397743 DOI: 10.1093/brain/awae323] [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: 06/17/2024] [Revised: 08/15/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024] Open
Abstract
Muscle diseases cover a diverse group of disorders that, in most cases, are hereditary. The rarity of the individual muscle diseases provides a challenge for researchers when wanting to establish natural history of the conditions and when trying to develop diagnostic tools, therapies, and outcome measures to evaluate disease progression. With emerging molecular therapies in many genetic muscle diseases, as well as biological therapies for the immune-mediated diseases, biological biomarkers play an important role in both drug development and evaluation. In this review, we focus on the role of biological biomarkers in muscle diseases and discuss their utility as surrogate end points in therapeutic trials. We categorize these as either (i) disease unspecific markers; (ii) markers of specific pathways that may be used for more than one disease; or (iii) disease-specific markers. We also propose that evaluation of specific therapeutic interventions benefits from biological markers that match the intervention.
Collapse
Affiliation(s)
- Mads G Stemmerik
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Giorgio Tasca
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trusts, Newcastle Upon Tyne NE1 3BZ, UK
| | - Nils Erik Gilhus
- Department of Clinical Medicine, University of Bergen, 5007 Bergen, Norway
- Department of Neurology, Haukeland University Hospital, 5009 Bergen, Norway
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre and NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX1 3PT, UK
- Division of Child Neurology, Department of Pediatrics, Centre de Référence des Maladies Neuromusculaires, University Hospital Liège and University of Liège, 4000 Liège, Belgium
| | - Alex Vicino
- Nerve-Muscle Unit, Neurology Service, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - Lorenzo Maggi
- Neuroimmunology and Neuromuscular Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan MI, Italy
| | - Valeria Sansone
- The NEMO Clinical Center in Milan, Neurorehabilitation Unit, University of Milan- ERN for Neuromuscular Diseases, 20162 Milan MI, Italy
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
6
|
Nigam P, Fitzgerald KK, Scavina M, Tsuda T. Diverse Cardiac Phenotype of Becker Muscular Dystrophy: Under-Recognized Subclinical Cardiomyopathy Due to Partial Dystrophin Deficiency in a Contemporary Era. Pediatr Cardiol 2025; 46:267-278. [PMID: 38240762 DOI: 10.1007/s00246-023-03382-9] [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: 07/25/2023] [Accepted: 12/10/2023] [Indexed: 02/02/2025]
Abstract
Becker muscular dystrophy (BMD) is an X-linked recessive disorder responsible for mild skeletal muscle involvement and variable degree of cardiomyopathy. The characteristics of cardiac phenotype of BMD in childhood remain elusive. Clinical manifestations, genotype, serum biomarkers, and echocardiogram were retrospectively reviewed in BMD patients. Cardiac phenotype was classified into acute progressive (AP), chronic persistent (CP), and latent (L) groups based upon symptoms and echocardiographic findings. Twenty-five BMD patients were studied over 9.5 ± 2.5 years. Sixteen patients presented initially with variable degree of muscle weakness whereas 9 were asymptomatic. Three patients developed medically refractory heart failure by age 18 with progressive dilated cardiomyopathy (DCM) (AP). Six patients developed mild to moderate left ventricular (LV) systolic dysfunction with LV dilatation but remained asymptomatic (CP). Although 16 patients continued to show normal LV function (L), they demonstrated variable degrees of skeletal muscle involvement. The AP groups presented with significantly larger LV size and LV mass index (LVMI) at the initial encounter than groups CP or L, suggesting early myocardial remodeling predicts rapid disease progression. None presented with atrophic myocardial phenotype commonly observed in Duchenne muscular dystrophy (DMD). Wide availability of genetic testing has changed the scope of clinical presentation of BMD. Cardiomyopathy in BMD presents with a diverse clinical spectrum with variable progression of DCM where larger LV dimension and mass at the time of diagnosis may predict the progressiveness of cardiomyopathy.
Collapse
Affiliation(s)
- Priya Nigam
- Nemours Cardiac Center, Nemours Children's Hospital Delaware, 1600 Rockland Rd, Wilmington, DE, 19803, USA
- Children's Heart Program, University of Maryland Medical Center, Baltimore, MD, 21201, USA
| | - Kristi K Fitzgerald
- Nemours Cardiac Center, Nemours Children's Hospital Delaware, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| | - Mena Scavina
- Division of Neurology, Nemours Children's Hospital Delaware, 1600 Rockland Rd, Wilmington, DE, 19803, USA
| | - Takeshi Tsuda
- Nemours Cardiac Center, Nemours Children's Hospital Delaware, 1600 Rockland Rd, Wilmington, DE, 19803, USA.
- Department of Pediatrics, Sidney Kimmel Medical College at Thomas Jefferson University, 1015 Walnut St, Philadelphia, PA, 19107, USA.
| |
Collapse
|
7
|
Poyatos-García J, Soblechero-Martín P, Liquori A, López-Martínez A, Maestre P, González-Romero E, Vázquez-Manrique RP, Muelas N, García-García G, Ohana J, Arechavala-Gomeza V, Vílchez JJ. Deletion of exons 45 to 55 in the DMD gene: from the therapeutic perspective to the in vitro model. Skelet Muscle 2024; 14:21. [PMID: 39354597 PMCID: PMC11443720 DOI: 10.1186/s13395-024-00353-3] [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: 01/07/2024] [Accepted: 09/13/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Gene editing therapies in development for correcting out-of-frame DMD mutations in Duchenne muscular dystrophy aim to replicate benign spontaneous deletions. Deletion of 45-55 DMD exons (del45-55) was described in asymptomatic subjects, but recently serious skeletal and cardiac complications have been reported. Uncovering why a single mutation like del45-55 is able to induce diverse phenotypes and grades of severity may impact the strategies of emerging therapies. Cellular models are essential for this purpose, but their availability is compromised by scarce muscle biopsies. METHODS We introduced, as a proof-of-concept, using CRISPR-Cas9 edition, a del45-55 mimicking the intronic breakpoints harboured by a subset of patients of this form of dystrophinopathy (designing specific gRNAs), into a Duchenne patient's cell line. The edited cell line was characterized evaluating the dystrophin expression and the myogenic status. RESULTS Dystrophin expression was restored, and the myogenic defects were ameliorated in the edited myoblasts harbouring a specific del45-55. Besides confirming the potential of CRISPR-Cas9 to create tailored mutations (despite the low cleavage efficiency of our gRNAs) as a useful approach to generate in vitro models, we also generated an immortalized myoblast line derived from a patient with a specific del45-55. CONCLUSIONS Overall, we provide helpful resources to deepen into unknown factors responsible for DMD-pathophysiology.
Collapse
Affiliation(s)
- Javier Poyatos-García
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), CB23/07/00005, Madrid, Spain.
- University of Valencia, Valencia, Spain.
| | - Patricia Soblechero-Martín
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Alessandro Liquori
- Hematology Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
- Centre for Biomedical Network Research on Cancer (CIBERONC), CB16/12/00284, Madrid, Spain
| | - Andrea López-Martínez
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Pilar Maestre
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Elisa González-Romero
- Hematology Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
| | - Rafael P Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Health Research Institute Hospital La Fe, Valencia, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U755, CB06/07/1030, Madrid, Spain
- Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - Nuria Muelas
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain
- Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO- NMD), Hospital Universitari I Politècnic La Fe, Valencia, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, Madrid, Spain
| | - Gema García-García
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Health Research Institute Hospital La Fe, Valencia, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U755, CB06/07/1030, Madrid, Spain
- Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - Jessica Ohana
- Centre de Recherche en Myologie, Sorbonne Université, INSERM, Institut de Myologie, Paris, 75013, France
| | - Virginia Arechavala-Gomeza
- Nucleic Acid Therapeutics for Rare Disorders (NAT-RD), Biobizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Juan J Vílchez
- Neuromuscular and Ataxias Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Valencia, Spain.
- University of Valencia, Valencia, Spain.
- Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO- NMD), Hospital Universitari I Politècnic La Fe, Valencia, Spain.
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), U763, CB06/05/0091, Madrid, Spain.
| |
Collapse
|
8
|
Stirm M, Klymiuk N, Nagashima H, Kupatt C, Wolf E. Pig models for translational Duchenne muscular dystrophy research. Trends Mol Med 2024; 30:950-964. [PMID: 38749865 DOI: 10.1016/j.molmed.2024.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 10/12/2024]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the X-linked DMD gene, resulting in the absence of dystrophin, progressive muscle degeneration, and heart failure. Genetically tailored pig models resembling human DMD mutations recapitulate the biochemical, clinical, and pathological hallmarks of DMD with an accelerated disease progression compared to human patients. DMD pigs have been used to evaluate therapeutic concepts such as gene editing to reframe a disrupted DMD reading frame or the delivery of artificial chromosome vectors carrying the complete DMD gene. Moreover, DMD pigs have been instrumental in validating new diagnostic modalities such as multispectral optoacoustic tomography (MSOT) for non-invasive monitoring of disease progression. DMD pigs may thus help to bridge the gap between proof-of-concept studies in cellular or rodent models and clinical studies in patients.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Large Animal Models in Cardiovascular Research, Internal Medical Department I, Technical University of Munich (TU Munich), 81675 Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa 214-8571, Japan
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, 81675 Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
9
|
Baylot V, Le TK, Taïeb D, Rocchi P, Colleaux L. Between hope and reality: treatment of genetic diseases through nucleic acid-based drugs. Commun Biol 2024; 7:489. [PMID: 38653753 PMCID: PMC11039704 DOI: 10.1038/s42003-024-06121-9] [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: 08/22/2023] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Rare diseases (RD) affect a small number of people compared to the general population and are mostly genetic in origin. The first clinical signs often appear at birth or in childhood, and patients endure high levels of pain and progressive loss of autonomy frequently associated with short life expectancy. Until recently, the low prevalence of RD and the gatekeeping delay in their diagnosis have long hampered research. The era of nucleic acid (NA)-based therapies has revolutionized the landscape of RD treatment and new hopes arise with the perspectives of disease-modifying drugs development as some NA-based therapies are now entering the clinical stage. Herein, we review NA-based drugs that were approved and are currently under investigation for the treatment of RD. We also discuss the recent structural improvements of NA-based therapeutics and delivery system, which overcome the main limitations in their market expansion and the current approaches that are developed to address the endosomal escape issue. We finally open the discussion on the ethical and societal issues that raise this new technology in terms of regulatory approval and sustainability of production.
Collapse
Affiliation(s)
- Virginie Baylot
- Aix Marseille Univ, CNRS, CINAM, ERL INSERM U 1326, CERIMED, Marseille, France.
| | - Thi Khanh Le
- Aix Marseille Univ, CNRS, CINAM, ERL INSERM U 1326, CERIMED, Marseille, France
| | - David Taïeb
- Aix Marseille Univ, CNRS, CINAM, ERL INSERM U 1326, CERIMED, Marseille, France
| | - Palma Rocchi
- Aix Marseille Univ, CNRS, CINAM, ERL INSERM U 1326, CERIMED, Marseille, France.
| | - Laurence Colleaux
- Aix Marseille Univ, CNRS, CINAM, ERL INSERM U 1326, CERIMED, Marseille, France
| |
Collapse
|
10
|
Fortunato F, Tonelli L, Farnè M, Selvatici R, Ferlini A. DMD deletions underlining mild dystrophinopathies: literature review highlights phenotype-related mutation clusters and provides insights about genetic mechanisms and prognosis. Front Neurol 2024; 14:1288721. [PMID: 38288333 PMCID: PMC10823016 DOI: 10.3389/fneur.2023.1288721] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 01/31/2024] Open
Abstract
DMD gene pathogenic variations cause a spectrum of phenotypes, ranging from severe Duchenne muscular dystrophy, the Becker milder cases, the intermediate or very mild muscle phenotypes invariably characterized by high CK, and the ultrarare fully-asymptomatic cases. Besides these phenotypes, X-linked dilated cardiomyopathy is also caused by DMD mutations. Males carrying DMD deletions with absent or very mild phenotypes have been sparsely described. We performed a horizon scan on public datasets to enroll males with the above phenotypes and carrying DMD deletions to delineate myopathic genotype-phenotype relationships. We inventoried 81 males, who were divided into the following clinical categorization: fully-asymptomatic males aged >43 years (A, N = 22); isolated hyperCKemia (CK, N = 35); and mild weakness (any age) with or without high CK (WCK, N = 24). In all cases, deleted intervals were exons 2 to 55, and no downstream exons were ever involved, apart from an exon 78 deletion in a WCK patient. All deletions were in-frame apart from the known exception to the rule of exon 2 and exon 78. We correlated the mild phenotypes (A and CK) to deleted exons, intronic breakpoints, exon-exon junctions, 3' isoforms rule, and protein epitopes, and we found that some genetic profiles are exclusively/mainly occurring in A/CK phenotypes, suggesting they are compatible with a quasi-normal muscular performance. We discussed diverse pathogenic mechanisms that may contribute to mild dystrophinopathic phenotypes, and we tried to address some "critical" genetic configurations or exon content needed to preserve a semi-functional DMD gene.
Collapse
Affiliation(s)
| | | | | | | | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
11
|
Nambu Y, Shirakawa T, Osawa K, Nishio H, Nozu K, Matsuo M, Awano H. Brothers with Becker muscular dystrophy show discordance in skeletal muscle computed tomography findings: A case report. SAGE Open Med Case Rep 2024; 12:2050313X231221436. [PMID: 38187815 PMCID: PMC10768573 DOI: 10.1177/2050313x231221436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/29/2023] [Indexed: 01/09/2024] Open
Abstract
Becker muscular dystrophy is caused by DMD mutations and is characterized by progressive muscle atrophy. The wide variations observed in muscle atrophy progression in Becker muscular dystrophy are considered multifactorial, including differences in mutations and environmental factors. In this case, two brothers, aged 2 and 3 years, had the identical DMD mutation, confirming their Becker muscular dystrophy diagnosis. They began using handrails when ascending and descending stairs at the age of 16 due to progressive muscular weakness. Over an 18-year follow-up, the older brother consistently had high serum creatine kinase levels, significantly over median levels. Muscle computed tomography finings revealed that the older brother's gluteus maximus and vastus femoris cross-sectional areas were only half and one-third of the younger brother's, respectively. The mean computed tomography values of gluteus maximus and vastus femoris were significantly lower in the older brother. Our report suggests that muscle atrophy in Becker muscular dystrophy cannot be solely explained by dystrophin mutation or environmental factors.
Collapse
Affiliation(s)
- Yoshinori Nambu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Taku Shirakawa
- Faculty of Health Sciences, Department of Medical Technology, Kobe Tokiwa University, Kobe, Japan
| | - Kayo Osawa
- Faculty of Health Sciences, Department of Medical Technology, Kobe Tokiwa University, Kobe, Japan
| | - Hisahide Nishio
- Faculty of Rehabilitation, Research Center for Locomotion Biology, Kobe Gakuin University, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masafumi Matsuo
- Faculty of Health Sciences, Department of Medical Technology, Kobe Tokiwa University, Kobe, Japan
| | - Hiroyuki Awano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
- Organization for Research Initiative and Promotion, Tottori University, Yonago, Japan
| |
Collapse
|
12
|
Saad FA, Saad JF, Siciliano G, Merlini L, Angelini C. Duchenne Muscular Dystrophy Gene Therapy. Curr Gene Ther 2024; 24:17-28. [PMID: 36411557 DOI: 10.2174/1566523223666221118160932] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022]
Abstract
Duchenne and Becker muscular dystrophies are allelic X-linked recessive neuromuscular diseases affecting both skeletal and cardiac muscles. Therefore, owing to their single X chromosome, the affected boys receive pathogenic gene mutations from their unknowing carrier mothers. Current pharmacological drugs are palliative that address the symptoms of the disease rather than the genetic cause imbedded in the Dystrophin gene DNA sequence. Therefore, alternative therapies like gene drugs that could address the genetic cause of the disease at its root are crucial, which include gene transfer/implantation, exon skipping, and gene editing. Presently, it is possible through genetic reprogramming to engineer AAV vectors to deliver certain therapeutic cargos specifically to muscle or other organs regardless of their serotype. Similarly, it is possible to direct the biogenesis of exosomes to carry gene editing constituents or certain therapeutic cargos to specific tissue or cell type like brain and muscle. While autologous exosomes are immunologically inert, it is possible to camouflage AAV capsids, and lipid nanoparticles to evade the immune system recognition. In this review, we highlight current opportunities for Duchenne muscular dystrophy gene therapy, which has been known thus far as an incurable genetic disease. This article is a part of Gene Therapy of Rare Genetic Diseases thematic issue.
Collapse
Affiliation(s)
- Fawzy A Saad
- Department of Biology, Padua University School of Medicine, Via Trieste 75, Padova 35121, Italy
- Department of Gene Therapy, Saad Pharmaceuticals, Tornimäe 7-26, Tallinn, 10145, Estonia
| | - Jasen F Saad
- Department of Gene Therapy, Saad Pharmaceuticals, Tornimäe 7-26, Tallinn, 10145, Estonia
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, Pisa University School of Medicine, Pisa, Italy
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Sciences, Bologna University School of Medicine, 40126 Bologna, Italy
| | - Corrado Angelini
- Department Neurosciences, Padova University School of Medicine, Padova, Italy
| |
Collapse
|
13
|
Clemens PR, Gordish-Dressman H, Niizawa G, Gorni K, Guglieri M, Connolly AM, Wicklund M, Bertorini T, Mah J, Thangarajh M, Smith EC, Kuntz NL, McDonald CM, Henricson E, Upadhyayula S, Byrne B, Manousakis G, Harper A, Iannaccone S, Dang UJ. Findings from the Longitudinal CINRG Becker Natural History Study. J Neuromuscul Dis 2024; 11:201-212. [PMID: 37980682 PMCID: PMC10789327 DOI: 10.3233/jnd-230178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Becker muscular dystrophy is an X-linked, genetic disorder causing progressive degeneration of skeletal and cardiac muscle, with a widely variable phenotype. OBJECTIVE A 3-year, longitudinal, prospective dataset contributed by patients with confirmed Becker muscular dystrophy was analyzed to characterize the natural history of this disorder. A better understanding of the natural history is crucial to rigorous therapeutic trials. METHODS A cohort of 83 patients with Becker muscular dystrophy (5-75 years at baseline) were followed for up to 3 years with annual assessments. Muscle and pulmonary function outcomes were analyzed herein. Age-stratified statistical analysis and modeling were conducted to analyze cross-sectional data, time-to-event data, and longitudinal data to characterize these clinical outcomes. RESULTS Deletion mutations of dystrophin exons 45-47 or 45-48 were most common. Subgroup analysis showed greater pairwise association between motor outcomes at baseline than association between these outcomes and age. Stronger correlations between outcomes for adults than for those under 18 years were also observed. Using cross-sectional binning analysis, a ceiling effect was seen for North Star Ambulatory Assessment but not for other functional outcomes. Longitudinal analysis showed a decline in percentage predicted forced vital capacity over the life span. There was relative stability or improved median function for motor functional outcomes through childhood and adolescence and decreasing function with age thereafter. CONCLUSIONS There is variable progression of outcomes resulting in significant heterogeneity of the clinical phenotype of Becker muscular dystrophy. Disease progression is largely manifest in adulthood. There are implications for clinical trial design revealed by this longitudinal analysis of a Becker natural history dataset.
Collapse
Affiliation(s)
| | | | | | | | - Michela Guglieri
- Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Matthew Wicklund
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | | | - Jean Mah
- Cumming School of Medicine, University of Calgary, Calgary, Canada
| | | | | | | | | | | | | | | | | | - Amy Harper
- Virginia Commonwealth University, Richmond, VA, USA
| | - Susan Iannaccone
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
14
|
Ricci G, Govoni A, Torri F, Astrea G, Buchignani B, Marinella G, Battini R, Manca ML, Castiglione V, Giannoni A, Emdin M, Siciliano G. Characterization of Phenotypic Variability in Becker Muscular Dystrophy for Clinical Practice and Towards Trial Readiness: A Two-Years Follow up Study. J Neuromuscul Dis 2024; 11:375-387. [PMID: 38189759 DOI: 10.3233/jnd-221513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Background Becker muscular dystrophy (BMD) is a dystrophinopathy due to in-frame mutations in the dystrophin gene (DMD) which determines a reduction of dystrophin at muscle level. BMD has a wide spectrum of clinical variability with different degrees of disability. Studies of natural history are needed also in view of up-coming clinical trials. Objectives From an initial cohort of 32 BMD adult subjects, we present a detailed phenotypic characterization of 28 patients, then providing a description of their clinical natural history over the course of 12 months for 18 and 24 months for 13 of them. Methods Each patient has been genetically characterized. Baseline, and 1-year and 2 years assessments included North Star Ambulatory Assessment (NSAA), timed function tests (time to climb and descend four stairs), 6-minute walk test (6MWT), Walton and Gardner-Medwin Scale and Medical Research Council (MRC) scale. Muscle magnetic resonance imaging (MRI) was acquired at baseline and in a subgroup of 9 patients after 24 months. Data on cardiac function (electrocardiogram, echocardiogram, and cardiac MRI) were also collected. Results and conclusions Among the clinical heterogeneity, a more severe involvement is often observed in patients with 45-X del, with a disease progression over two years. The 6MWT appears sensitive to detect modification from baseline during follow up while no variation was observed by MRC testing. Muscle MRI of the lower limbs correlates with clinical parameters.Our study further highlights how the phenotypic variability of BMD adult patients makes it difficult to describe an uniform course and substantiates the need to identify predictive parameters and biomarkers to stratify patients.
Collapse
Affiliation(s)
- Giulia Ricci
- Department of Clinical and Experimental Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
| | - Alessandra Govoni
- Neuromuscular and Rare Disease Unit, La Fondazione IRCCS Ca' Granda Ospedale Maggiore di Milano Policlinico, Milano, Italy
| | - Francesca Torri
- Department of Clinical and Experimental Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
| | - Guja Astrea
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Bianca Buchignani
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
- Department of Translational Research and of New Surgical and Medical Technologies Pisa University, Pisa, Italy
| | - Gemma Marinella
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Roberta Battini
- Department of Clinical and Experimental Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
- Department of Developmental Neuroscience, IRCCS Stella Maris, Calambrone, Pisa, Italy
| | - Maria Laura Manca
- Department of Clinical and Experimental Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
- Department of Mathematics, University of Pisa, Pisa, Italy
| | - Vincenzo Castiglione
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Giannoni
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa, Italy
- Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, Neurological Clinic, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Eslahi A, Alizadeh F, Avan A, Ferns GA, Moghbeli M, Reza Abbaszadegan M, Mojarrad M. New advancements in CRISPR based gene therapy of Duchenne muscular dystrophy. Gene 2023; 867:147358. [PMID: 36914142 DOI: 10.1016/j.gene.2023.147358] [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: 11/25/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Duchenne muscular dystrophy (DMD) is caused by the dystrophin gene mutations and is one of the most common and lethal human hereditary disorders. A novel therapeutic approach using CRISPR technology has gained attention in the treatment of DMD. Gene replacement strategies are being proposed as a promising therapeutic option to compensate the loss of function mutations. Although, the large size of the dystrophin gene and the limitations of the existing gene replacement approach, could mean the gene delivery of shortened versions of dystrophin such as midystrophin and microdystrophins. There are also other approaches: including Targeted removal of dystrophin exons to restore the reading-frame; Dual sgRNA-directed DMD exon deletion, CRISPR-SKIP strategy; reframing of dystrophin using Prime Editing technology; exon removal using twin prime technology; TransCRISTI technology to targeted exon integration into dystrophin gene. Here we provide an overview of recent progresses in dystrophin gene editing using updated versions of CRISPR to introduce novel opportunities in DMD gene therapy. Overall, the novel CRISPR based technologies are improving and expanding to allow the application of more precise gene editing for the treatment of DMD.
Collapse
Affiliation(s)
- Atieh Eslahi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzaneh Alizadeh
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Mojarrad
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Genetic Center of Khorasan Razavi, Mashhad, Iran.
| |
Collapse
|
16
|
Boehler JF, Brown KJ, Beatka M, Gonzalez JP, Donisa Dreghici R, Soustek-Kramer M, McGonigle S, Ganot A, Palmer T, Lowie C, Chamberlain JS, Lawlor MW, Morris CA. Clinical potential of microdystrophin as a surrogate endpoint. Neuromuscul Disord 2023; 33:40-49. [PMID: 36575103 DOI: 10.1016/j.nmd.2022.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/28/2022] [Accepted: 12/13/2022] [Indexed: 12/15/2022]
Abstract
Accelerated approval based on a likely surrogate endpoint can be life-changing for patients suffering from a rare progressive disease with unmet medical need, as it substantially hastens access to potentially lifesaving therapies. In one such example, antisense morpholinos were approved to treat Duchenne muscular dystrophy (DMD) based on measurement of shortened dystrophin in skeletal muscle biopsies as a surrogate biomarker. New, promising therapeutics for DMD include AAV gene therapy to restore another form of dystrophin termed mini- or microdystrophin. AAV-microdystrophins are currently in clinical trials but have yet to be accepted by regulatory agencies as reasonably likely surrogate endpoints. To evaluate microdystrophin expression as a reasonably likely surrogate endpoint for DMD, this review highlights dystrophin biology in the context of functional and clinical benefit to support the argument that microdystrophin proteins have a high probability of providing clinical benefit based on their rational design. Unlike exon-skipping based strategies, the approach of rational design allows for functional capabilities (i.e. quality) of the protein to be maximized with every patient receiving the same optimized microdystrophin. Therefore, the presence of rationally designed microdystrophin in a muscle biopsy is likely to predict clinical benefit and is consequently a strong candidate for a surrogate endpoint analysis to support accelerated approval.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Kristy J Brown
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Margaret Beatka
- Diverge TSL, 247 Freshwater Way Suite 610, Milwaukee, WI 53204, United States
| | - J Patrick Gonzalez
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | | | | | - Sharon McGonigle
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Annie Ganot
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Timothy Palmer
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Caitlin Lowie
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, United States
| | - Michael W Lawlor
- Diverge TSL, 247 Freshwater Way Suite 610, Milwaukee, WI 53204, United States
| | - Carl A Morris
- Solid Biosciences, 500 Rutherford Avenue 3rd Floor, Boston, MA 02129, United States.
| |
Collapse
|
17
|
Hanson B, Stenler S, Ahlskog N, Chwalenia K, Svrzikapa N, Coenen-Stass AM, Weinberg MS, Wood MJ, Roberts TC. Non-uniform dystrophin re-expression after CRISPR-mediated exon excision in the dystrophin/utrophin double-knockout mouse model of DMD. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:379-397. [PMID: 36420212 PMCID: PMC9664411 DOI: 10.1016/j.omtn.2022.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most prevalent inherited myopathy affecting children, caused by genetic loss of the gene encoding the dystrophin protein. Here we have investigated the use of the Staphylococcus aureus CRISPR-Cas9 system and a double-cut strategy, delivered using a pair of adeno-associated virus serotype 9 (AAV9) vectors, for dystrophin restoration in the severely affected dystrophin/utrophin double-knockout (dKO) mouse. Single guide RNAs were designed to excise Dmd exon 23, with flanking intronic regions repaired by non-homologous end joining. Exon 23 deletion was confirmed at the DNA level by PCR and Sanger sequencing, and at the RNA level by RT-qPCR. Restoration of dystrophin protein expression was demonstrated by western blot and immunofluorescence staining in mice treated via either intraperitoneal or intravenous routes of delivery. Dystrophin restoration was most effective in the diaphragm, where a maximum of 5.7% of wild-type dystrophin expression was observed. CRISPR treatment was insufficient to extend lifespan in the dKO mouse, and dystrophin was expressed in a within-fiber patchy manner in skeletal muscle tissues. Further analysis revealed a plethora of non-productive DNA repair events, including AAV genome integration at the CRISPR cut sites. This study highlights potential challenges for the successful development of CRISPR therapies in the context of DMD.
Collapse
Affiliation(s)
- Britt Hanson
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Sofia Stenler
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Nina Ahlskog
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Katarzyna Chwalenia
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
| | - Nenad Svrzikapa
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- Wave Life Sciences Ltd., Cambridge, MA 02138, USA
| | - Anna M.L. Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
| | - Marc S. Weinberg
- Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, University of the Witwatersrand Medical School, WITS 2050, South Africa
- Asklepios BioPharmaceutical, Inc., Research Triangle Park, NC 27709, USA
| | - Matthew J.A. Wood
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX1 3QX, UK
| | - Thomas C. Roberts
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK
- Institute of Developmental and Regenerative Medicine, University of Oxford, IMS-Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, Headington, Oxford OX3 7TY, UK
- MDUK Oxford Neuromuscular Centre, South Parks Road, Oxford OX1 3QX, UK
| |
Collapse
|
18
|
Poyatos‐García J, Martí P, Liquori A, Muelas N, Pitarch I, Martinez‐Dolz L, Rodríguez B, Gonzalez‐Quereda L, Damiá M, Aller E, Selva‐Gimenez M, Vilchez R, Diaz‐Manera J, Alonso‐Pérez J, Barcena JE, Jauregui A, Gámez J, Aladrén JA, Fernández A, Montolio M, Azorin I, Hervas D, Casasús A, Nieto M, Gallano P, Sevilla T, Vilchez JJ. Dystrophinopathy Phenotypes and Modifying Factors in DMD Exon 45-55 Deletion. Ann Neurol 2022; 92:793-806. [PMID: 35897138 PMCID: PMC9825930 DOI: 10.1002/ana.26461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) exon 45-55 deletion (del45-55) has been postulated as a model that could treat up to 60% of DMD patients, but the associated clinical variability and complications require clarification. We aimed to understand the phenotypes and potential modifying factors of this dystrophinopathy subset. METHODS This cross-sectional, multicenter cohort study applied clinical and functional evaluation. Next generation sequencing was employed to identify intronic breakpoints and their impact on the Dp140 promotor, intronic long noncoding RNA, and regulatory splicing sequences. DMD modifiers (SPP1, LTBP4, ACTN3) and concomitant mutations were also assessed. Haplotypes were built using DMD single nucleotide polymorphisms. Dystrophin expression was evaluated via immunostaining, Western blotting, reverse transcription polymerase chain reaction (PCR), and droplet digital PCR in 9 muscle biopsies. RESULTS The series comprised 57 subjects (23 index) expressing Becker phenotype (28%), isolated cardiopathy (19%), and asymptomatic features (53%). Cognitive impairment occurred in 90% of children. Patients were classified according to 10 distinct index-case breakpoints; 4 of them were recurrent due to founder events. A specific breakpoint (D5) was associated with severity, but no significant effect was appreciated due to the changes in intronic sequences. All biopsies showed dystrophin expression of >67% and traces of alternative del45-57 transcript that were not deemed pathogenically relevant. Only the LTBP4 haplotype appeared associated the presence of cardiopathy among the explored extragenic factors. INTERPRETATION We confirmed that del45-55 segregates a high proportion of benign phenotypes, severe cases, and isolated cardiac and cognitive presentations. Although some influence of the intronic breakpoint position and the LTBP4 modifier may exist, the pathomechanisms responsible for the phenotypic variability remain largely unresolved. ANN NEUROL 2022;92:793-806.
Collapse
Affiliation(s)
- Javier Poyatos‐García
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Pilar Martí
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Alessandro Liquori
- Hematology Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Cancer (CIBERONC); CB16/12/00284MadridSpain
| | - Nuria Muelas
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain,Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Universitary and Polytechnic La Fe HospitalValenciaSpain
| | - Inmaculada Pitarch
- Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Universitary and Polytechnic La Fe HospitalValenciaSpain,Neuropediatric DepartmentUniversitary and Polytechnic La Fe HospitalValenciaSpain
| | - Luis Martinez‐Dolz
- Cardiology DepartmentUniversity and Polytechnic La Fe Hospital, IIS La FeValenciaSpain,Centre for Biomedical Network Research on Cardiovascular Diseases (CIBERCV)ValenciaSpain
| | - Benjamin Rodríguez
- Genetics DepartmentIIB Sant Pau, Hospital of Sant PauBarcelonaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER)U705, U745, CB06/07/0011BarcelonaSpain
| | - Lidia Gonzalez‐Quereda
- Genetics DepartmentIIB Sant Pau, Hospital of Sant PauBarcelonaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER)U705, U745, CB06/07/0011BarcelonaSpain
| | - Maria Damiá
- Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Universitary and Polytechnic La Fe HospitalValenciaSpain,Neuropediatric DepartmentUniversitary and Polytechnic La Fe HospitalValenciaSpain
| | - Elena Aller
- Genetics UnitUniversitary and Polytechnic La Fe HospitalValenciaSpain
| | - Marta Selva‐Gimenez
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Roger Vilchez
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Jordi Diaz‐Manera
- Neuromuscular Disorders Unit, Neurology Department, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Hospital of Sant PauBarcelonaSpain,Autonomous University of BarcelonaBarcelonaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER)U762, CB06/05/0030BarcelonaSpain
| | - Jorge Alonso‐Pérez
- Neuromuscular Disorders Unit, Neurology Department, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Hospital of Sant PauBarcelonaSpain,Autonomous University of BarcelonaBarcelonaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER)U762, CB06/05/0030BarcelonaSpain
| | - José Eulalio Barcena
- Neuromuscular Section, Neurology ServiceCruces University HospitalBarakaldoSpain
| | - Amaia Jauregui
- Neuromuscular Section, Neurology ServiceCruces University HospitalBarakaldoSpain
| | - Josep Gámez
- Autonomous University of BarcelonaBarcelonaSpain,Neurology Department, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)GMA ClinicBarcelonaSpain
| | | | | | - Marisol Montolio
- Duchenne Parent Project SpainMadridSpain,Department of Cell Biology, Physiology, and Immunology, Faculty of BiologyBarcelonaSpain
| | - Inmaculada Azorin
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - David Hervas
- Department of Applied Statistics and Operations Research, and QualityPolytechnic University of ValenciaValenciaSpain
| | - Ana Casasús
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Marisa Nieto
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain
| | - Pia Gallano
- Genetics DepartmentIIB Sant Pau, Hospital of Sant PauBarcelonaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER)U705, U745, CB06/07/0011BarcelonaSpain
| | - Teresa Sevilla
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain,Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Universitary and Polytechnic La Fe HospitalValenciaSpain,Department of MedicineUniversity of ValenciaValenciaSpain
| | - Juan Jesus Vilchez
- Neuromuscular and Ataxias Research GroupHealth Research Institute Hospital La Fe (IIS La Fe)ValenciaSpain,Centre for Biomedical Network Research on Rare Diseases (CIBERER); U763, CB06/05/0091ValenciaSpain,Neuromuscular Referral Center, European Reference Network on Rare Neuromuscular Diseases (ERN EURO‐NMD)Universitary and Polytechnic La Fe HospitalValenciaSpain,Department of MedicineUniversity of ValenciaValenciaSpain
| |
Collapse
|
19
|
Williams LA, Gerber DJ, Elder A, Tseng WC, Baru V, Delaney-Busch N, Ambrosi C, Mahimkar G, Joshi V, Shah H, Harikrishnan K, Upadhyay H, Rajendran SH, Dhandapani A, Meier J, Ryan SJ, Lewarch C, Black L, Douville J, Cinquino S, Legakis H, Nalbach K, Behrends C, Sato A, Galluzzi L, Yu TW, Brown D, Agrawal S, Margulies D, Kopin A, Dempsey GT. Developing antisense oligonucleotides for a TECPR2 mutation-induced, ultra-rare neurological disorder using patient-derived cellular models. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:189-203. [PMID: 35860385 PMCID: PMC9287140 DOI: 10.1016/j.omtn.2022.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 06/17/2022] [Indexed: 11/16/2022]
Abstract
Mutations in the TECPR2 gene are the cause of an ultra-rare neurological disorder characterized by intellectual disability, impaired speech, motor delay, and hypotonia evolving to spasticity, central sleep apnea, and premature death (SPG49 or HSAN9; OMIM: 615031). Little is known about the biological function of TECPR2, and there are currently no available disease-modifying therapies for this disease. Here we describe implementation of an antisense oligonucleotide (ASO) exon-skipping strategy targeting TECPR2 c.1319delT (p.Leu440Argfs∗19), a pathogenic variant that results in a premature stop codon within TECPR2 exon 8. We used patient-derived fibroblasts and induced pluripotent stem cell (iPSC)-derived neurons homozygous for the p.Leu440Argfs∗19 mutation to model the disease in vitro. Both patient-derived fibroblasts and neurons showed lack of TECPR2 protein expression. We designed and screened ASOs targeting sequences across the TECPR2 exon 8 region to identify molecules that induce exon 8 skipping and thereby remove the premature stop signal. TECPR2 exon 8 skipping restored in-frame expression of a TECPR2 protein variant (TECPR2ΔEx8) containing 1,300 of 1,411 amino acids. Optimization of ASO sequences generated a lead candidate (ASO-005-02) with ∼27 nM potency in patient-derived fibroblasts. To examine potential functional rescue induced by ASO-005-02, we used iPSC-derived neurons to analyze the neuronal localization of TECPR2ΔEx8 and showed that this form of TECPR2 retains the distinct, punctate neuronal expression pattern of full-length TECPR2. Finally, ASO-005-02 had an acceptable tolerability profile in vivo following a single 20-mg intrathecal dose in cynomolgus monkeys, showing some transient non-adverse behavioral effects with no correlating histopathology. Broad distribution of ASO-005-02 and induction of TECPR2 exon 8 skipping was detected in multiple central nervous system (CNS) tissues, supporting the potential utility of this therapeutic strategy for a subset of patients suffering from this rare disease.
Collapse
Affiliation(s)
- Luis A Williams
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - David J Gerber
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Amy Elder
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Wei Chou Tseng
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Valeriya Baru
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | | | | | - Gauri Mahimkar
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Vaibhav Joshi
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Himali Shah
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | | | - Hansini Upadhyay
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | | | | | - Joshua Meier
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Steven J Ryan
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Caitlin Lewarch
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Lauren Black
- Charles River Laboratories, Montreal, QC, Canada
| | | | | | | | - Karsten Nalbach
- Munich Cluster for Systems Neurology, Ludwig-Maximilians-Universität München, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Ludwig-Maximilians-Universität München, Germany
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Timothy W Yu
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Duncan Brown
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Sudhir Agrawal
- University of Massachusetts Medical School, Department of Medicine, Worcester, MA 01655, USA.,Arnay Sciences LLC, Shrewsbury, MA 01545, USA
| | - David Margulies
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| | - Alan Kopin
- Tufts University School of Medicine, Boston, MA, USA.,Luke Heller TECPR2 Foundation, Swampscott, MA, USA
| | - Graham T Dempsey
- Q-State Biosciences, 179 Sidney Street, Cambridge, MA 02139, USA
| |
Collapse
|
20
|
Deng J, Zhang J, Shi K, Liu Z. Drug development progress in duchenne muscular dystrophy. Front Pharmacol 2022; 13:950651. [PMID: 35935842 PMCID: PMC9353054 DOI: 10.3389/fphar.2022.950651] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe, progressive, and incurable X-linked disorder caused by mutations in the dystrophin gene. Patients with DMD have an absence of functional dystrophin protein, which results in chronic damage of muscle fibers during contraction, thus leading to deterioration of muscle quality and loss of muscle mass over time. Although there is currently no cure for DMD, improvements in treatment care and management could delay disease progression and improve quality of life, thereby prolonging life expectancy for these patients. Furthermore, active research efforts are ongoing to develop therapeutic strategies that target dystrophin deficiency, such as gene replacement therapies, exon skipping, and readthrough therapy, as well as strategies that target secondary pathology of DMD, such as novel anti-inflammatory compounds, myostatin inhibitors, and cardioprotective compounds. Furthermore, longitudinal modeling approaches have been used to characterize the progression of MRI and functional endpoints for predictive purposes to inform Go/No Go decisions in drug development. This review showcases approved drugs or drug candidates along their development paths and also provides information on primary endpoints and enrollment size of Ph2/3 and Ph3 trials in the DMD space.
Collapse
Affiliation(s)
- Jiexin Deng
- School of Nursing and Health, Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| | - Junshi Zhang
- Department of Neurology, Huaihe Hospital of Henan University, Kaifeng, China
| | - Keli Shi
- School of Medicine, Henan University, Kaifeng, China
| | - Zhigang Liu
- Department of Orthopedics, First Affiliated Hospital of Henan University, Kaifeng, China
- *Correspondence: Jiexin Deng, ; Zhigang Liu,
| |
Collapse
|
21
|
Barp A, Carraro E, Goggi G, Lizio A, Zanolini A, Messina C, Perego S, Verdelli C, Lombardi G, Sansone VA, Corbetta S. Body composition and myokines in a cohort of patients with Becker muscular dystrophy. Muscle Nerve 2022; 66:63-70. [PMID: 35474226 PMCID: PMC9321020 DOI: 10.1002/mus.27565] [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] [Received: 10/20/2021] [Revised: 04/19/2022] [Accepted: 04/23/2022] [Indexed: 11/13/2022]
Abstract
Introduction/Aims Becker muscular dystrophy (BMD) is an X‐linked disease leading to muscle wasting and weakness. The decrease in lean body mass (LBM) in Duchenne muscular dystrophy, has shown correlation with loss of muscle function and bone density (BD). Myokines (including irisin) are hormones secreted by skeletal muscle that allow crosstalk between muscle and bone. The present study analyzed body composition and circulating myokine levels in a cohort of BMD patients; moreover, the association between dual energy X‐ray absorptiometry (DXA) parameters, functional motor assessments, and myokine levels was investigated. Methods All patients underwent DXA, blood samples for myokine assays, and functional motor assessments. A group of healthy controls (HCs) was also included. Results Thirty BMD patients, median age at evaluation 36.0 y [26.0–41.0], were included. Twenty‐nine patients underwent whole‐body DXA. Median value of total body Z‐score was −0.70. The prevalence of low skeletal muscle mass defined as appendicular skeletal muscle mass index (ASMMI) < 7.59 kg/m2 was 83%. Irisin levels were significantly lower in BMD compared to HCs (p = .03). All DXA parameters showed significant correlation with the functional motor assessments, in particular the h2‐standardized lean mass lower limb index (p = .0006); h2‐standardized total fat mass showed negative correlations with North Star Ambulatory Assessment and 6 min walk test (p = .03). Discussion DXA is a useful tool to evaluate body composition in BMD patients; the decrease in BD and LBM is associated with a reduction of motor function in BMD.
Collapse
Affiliation(s)
- Andrea Barp
- Neurorehabilitation Unit, NeMO Clinical Center, University of Milan, Milan, Italy
| | - Elena Carraro
- Neurorehabilitation Unit, NeMO Clinical Center, University of Milan, Milan, Italy
| | - Giovanni Goggi
- Endocrinology and Diabetology Service, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Andrea Lizio
- Neurorehabilitation Unit, NeMO Clinical Center, University of Milan, Milan, Italy
| | - Alice Zanolini
- Neurorehabilitation Unit, NeMO Clinical Center, University of Milan, Milan, Italy
| | | | - Silvia Perego
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Chiara Verdelli
- Laboratory of Experimental Endocrinology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Valeria Ada Sansone
- Neurorehabilitation Unit, NeMO Clinical Center, University of Milan, Milan, Italy
| | - Sabrina Corbetta
- Endocrinology and Diabetology Service, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Biomedical, Surgery and Dental Sciences, University of Milan, Milan, Italy
| |
Collapse
|
22
|
Ripolone M, Velardo D, Mondello S, Zanotti S, Magri F, Minuti E, Cazzaniga S, Fortunato F, Ciscato P, Tiberio F, Sciacco M, Moggio M, Bettica P, Comi GP. Muscle histological changes in a large cohort of patients affected with Becker muscular dystrophy. Acta Neuropathol Commun 2022; 10:48. [PMID: 35395784 PMCID: PMC8994373 DOI: 10.1186/s40478-022-01354-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/25/2022] [Indexed: 11/17/2022] Open
Abstract
Becker muscular dystrophy (BMD) is a severe X-linked muscle disease. Age of onset, clinical variability, speed of progression and affected tissues display wide variability, making a clinical trial design for drug development very complex. The histopathological changes in skeletal muscle tissue are central to the pathogenesis, but they have not been thoroughly elucidated yet. Here we analysed muscle biopsies from a large cohort of BMD patients, focusing our attention on the histopathological muscle parameters, as fibrosis, fatty replacement, fibre cross sectional area, necrosis, regenerating fibres, splitting fibres, internalized nuclei and dystrophy evaluation. We correlated histological parameters with both demographic features and clinical functional evaluations. The most interesting results of our study are the accurate quantification of fibroadipose tissue replacement and the identification of some histopathological aspects that well correlate with clinical performances. Through correlation analysis, we divided our patients into three clusters with well-defined histological and clinical features. In conclusion, this is the first study that analyses in detail the histological characteristics of muscle biopsies in a large cohort of BMD patients, correlating them to a functional impairment. The collection of these data help to better understand the histopathological progression of the disease and can be useful to validate any pharmacological trial in which the modification of muscle biopsy is utilized as outcome measure.
Collapse
|
23
|
Abstract
RNA-based therapeutics have entered the mainstream with seemingly limitless possibilities to treat all categories of neurological disease. Here, common RNA-based drug modalities such as antisense oligonucleotides, small interfering RNAs, RNA aptamers, RNA-based vaccines and mRNA drugs are reviewed highlighting their current and potential applications. Rapid progress has been made across rare genetic diseases and neurodegenerative disorders, but safe and effective delivery to the brain remains a significant challenge for many applications. The advent of individualized RNA-based therapies for ultra-rare diseases is discussed against the backdrop of the emergence of this field into more common conditions such as Alzheimer's disease and ischaemic stroke. There remains significant untapped potential in the use of RNA-based therapeutics for behavioural disorders and tumours of the central nervous system; coupled with the accelerated development expected over the next decade, the true potential of RNA-based therapeutics to transform the therapeutic landscape in neurology remains to be uncovered.
Collapse
Affiliation(s)
- Karen Anthony
- Centre for Physical Activity and Life Sciences, University of Northampton, Northampton, UK
| |
Collapse
|
24
|
López-Martínez A, Soblechero-Martín P, Arechavala-Gomeza V. Evaluation of Exon Skipping and Dystrophin Restoration in In Vitro Models of Duchenne Muscular Dystrophy. Methods Mol Biol 2022; 2434:217-233. [PMID: 35213020 PMCID: PMC9703204 DOI: 10.1007/978-1-0716-2010-6_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Several exon skipping antisense oligonucleotides (eteplirsen, golodirsen, viltolarsen, and casimersen) have been approved for the treatment of Duchenne muscular dystrophy, but many more are in development targeting an array of different DMD exons. Preclinical screening of the new oligonucleotide sequences is routinely performed using patient-derived cell cultures, and evaluation of their efficacy may be performed at RNA and/or protein level. While several methods to assess exon skipping and dystrophin expression in cell culture have been developed, the choice of methodology often depends on the availability of specific research equipment.In this chapter, we describe and indicate the relevant bibliography of all the methods that may be used in this evaluation and describe in detail the protocols routinely followed at our institution, one to evaluate the efficacy of skipping at RNA level (nested PCR) and the other the restoration of protein expression (myoblot ), which provide good results using equipment largely available to most research laboratories.
Collapse
Affiliation(s)
- Andrea López-Martínez
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Patricia Soblechero-Martín
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Osakidetza Basque Health Service, Bilbao-Basurto Integrated Health Organisation, Basurto University Hospital, Clinical Laboratory Service, Bilbao, Spain
| | - Virginia Arechavala-Gomeza
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
- Neuromuscular Disorders Research Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.
| |
Collapse
|
25
|
Ricci G, Bello L, Torri F, Schirinzi E, Pegoraro E, Siciliano G. Therapeutic opportunities and clinical outcome measures in Duchenne muscular dystrophy. Neurol Sci 2022; 43:625-633. [PMID: 35608735 PMCID: PMC9126754 DOI: 10.1007/s10072-022-06085-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/14/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a devastatingly severe genetic muscle disease characterized by childhood-onset muscle weakness, leading to loss of motor function and premature death due to respiratory and cardiac insufficiency. DISCUSSION In the following three and half decades, DMD kept its paradigmatic role in the field of muscle diseases, with first systematic description of disease progression with ad hoc outcome measures and the first attempts at correcting the disease-causing gene defect by several molecular targets. Clinical trials are critical for developing and evaluating new treatments for DMD. CONCLUSIONS In the last 20 years, research efforts converged in characterization of the disease mechanism and development of therapeutic strategies. Same effort needs to be dedicated to the development of outcome measures able to capture clinical benefit in clinical trials.
Collapse
Affiliation(s)
- Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Francesca Torri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Erika Schirinzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Gaina G, Vossen RHAM, Manole E, Plesca DA, Ionica E. Combining Protein Expression and Molecular Data Improves Mutation Characterization of Dystrophinopathies. Front Neurol 2021; 12:718396. [PMID: 34950096 PMCID: PMC8689184 DOI: 10.3389/fneur.2021.718396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Duchenne and Becker muscular dystrophy are X-linked recessive inherited disorders characterized by progressive weakness due to skeletal muscle degeneration. Different mutations in the DMD gene, which encodes for dystrophin protein, are responsible for these disorders. The aim of our study was to investigate the relationship between type, size, and location of the mutation that occurs in the DMD gene and their effect on dystrophin protein expression in a cohort of 40 male dystrophinopathy patients and nine females, possible carriers. We evaluated the expression of dystrophin by immunofluorescence and immunoblotting. The mutational spectrum of the DMD gene was established by MLPA for large copy number variants, followed by HRM analysis for point mutations and sequencing of samples with an abnormal melting profile. MLPA revealed 30 deletions (75%) and three duplications (7.5%). HRM analysis accounted for seven-point mutations (17.5%). We also report four novel small mutations (c. 8507G>T, c.3021delG, c.9563_9563+1insAGCATGTTTATGATACAGCA, c.7661-60T>A) in DMD gene. Our work shows that the DNA translational open reading frame and the location of the mutation both influence the expression of dystrophin and disease severity phenotype. The proposed algorithm used in this study demonstrates its accuracy for the characterization of dystrophinopathy patients.
Collapse
Affiliation(s)
- Gisela Gaina
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, Bucharest, Romania
- *Correspondence: Gisela Gaina ;
| | - Rolf H. A. M. Vossen
- Center for Human and Clinical Genetics, Leiden Genome Technology Center, Leiden, Netherlands
| | - Emilia Manole
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, Bucharest, Romania
- Colentina Clinical Hospital, Bucharest, Romania
| | - Doina Anca Plesca
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Clinical Pediatrics, Victor Gomoiu Children Clinical Hospital, Bucharest, Romania
| | - Elena Ionica
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
27
|
Sheikh O, Yokota T. Pharmacology and toxicology of eteplirsen and SRP-5051 for DMD exon 51 skipping: an update. Arch Toxicol 2021; 96:1-9. [PMID: 34797383 DOI: 10.1007/s00204-021-03184-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/25/2021] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) afflicts 1 in 5000 newborn males, leading to progressive muscle weakening and the loss of ambulation between the ages of 8 and 12. Typically, DMD patients pass away from heart failure or respiratory failure. Currently, there is no cure, though exon-skipping therapy including eteplirsen (brand name Exondys 51), a synthetic antisense oligonucleotide designed to skip exon 51 of the dystrophin gene, is considered especially promising. Applicable to approximately 14% of DMD patients, a phosphorodiamidate morpholino oligomer (PMO) antisense oligonucleotide eteplirsen received accelerated approval by the US Food and Drug Administration (FDA) in 2016. Throughout clinical trials, eteplirsen has been well tolerated by patients with no serious drug-related adverse events. The most common events observed are balance disorder, vomiting, and skin rash. Despite its safety and promise of functional benefits, eteplirsen remains controversial due to its low production of dystrophin. In addition, unmodified PMOs have limited efficacy in the heart. To address these concerns of efficacy, eteplirsen has been conjugated to a proprietary cell-penetrating peptide; the conjugate is called SRP-5051. Compared to eteplirsen, SRP-5051 aims to better prompt exon-skipping and dystrophin production but may have greater toxicity concerns. This paper reviews and discusses the available information on the efficacy, safety, and tolerability data of eteplirsen and SRP-5051 from preclinical and clinical trials. Issues faced by eteplirsen and SRP-5051, including efficacy and safety, are identified. Lastly, the current state of eteplirsen and exon-skipping therapy in general as a strategy for the treatment of DMD are discussed.
Collapse
Affiliation(s)
- Omar Sheikh
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, T6G 2R3, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, T6G 2R3, Canada.
| |
Collapse
|
28
|
Soblechero-Martín P, Albiasu-Arteta E, Anton-Martinez A, de la Puente-Ovejero L, Garcia-Jimenez I, González-Iglesias G, Larrañaga-Aiestaran I, López-Martínez A, Poyatos-García J, Ruiz-Del-Yerro E, Gonzalez F, Arechavala-Gomeza V. Duchenne muscular dystrophy cell culture models created by CRISPR/Cas9 gene editing and their application in drug screening. Sci Rep 2021; 11:18188. [PMID: 34521928 PMCID: PMC8440673 DOI: 10.1038/s41598-021-97730-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/27/2021] [Indexed: 12/28/2022] Open
Abstract
Gene editing methods are an attractive therapeutic option for Duchenne muscular dystrophy, and they have an immediate application in the generation of research models. To generate myoblast cultures that could be useful in in vitro drug screening, we have optimised a CRISPR/Cas9 gene edition protocol. We have successfully used it in wild type immortalised myoblasts to delete exon 52 of the dystrophin gene, modelling a common Duchenne muscular dystrophy mutation; and in patient's immortalised cultures we have deleted an inhibitory microRNA target region of the utrophin UTR, leading to utrophin upregulation. We have characterised these cultures by demonstrating, respectively, inhibition of dystrophin expression and overexpression of utrophin, and evaluating the expression of myogenic factors (Myf5 and MyH3) and components of the dystrophin associated glycoprotein complex (α-sarcoglycan and β-dystroglycan). To demonstrate their use in the assessment of DMD treatments, we have performed exon skipping on the DMDΔ52-Model and have used the unedited DMD cultures/ DMD-UTRN-Model combo to assess utrophin overexpression after drug treatment. While the practical use of DMDΔ52-Model is limited to the validation to our gene editing protocol, DMD-UTRN-Model presents a possible therapeutic gene edition target as well as a useful positive control in the screening of utrophin overexpression drugs.
Collapse
Affiliation(s)
- Patricia Soblechero-Martín
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain.,Osakidetza Basque Health Service, Bilbao-Basurto Integrated Health Organisation, Basurto University Hospital, Clinical Laboratory Service, Bilbao, Spain
| | - Edurne Albiasu-Arteta
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | - Aina Anton-Martinez
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | | | - Iker Garcia-Jimenez
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | | | - Irene Larrañaga-Aiestaran
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | - Andrea López-Martínez
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | | | - Estíbaliz Ruiz-Del-Yerro
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | - Federico Gonzalez
- Pluripotent Stem Cells and Activation of Endogenous Tissue Programs for Organ Regeneration (PR Lab), Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Virginia Arechavala-Gomeza
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain. .,Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
29
|
Abstract
Duchenne muscular dystrophy (DMD) is a devastating, rare disease. While clinically described in the 19th century, the genetic foundation of DMD was not discovered until more than 100 years later. This genetic understanding opened the door to the development of genetic treatments for DMD. Over the course of the last 30 years, the research that supports this development has moved into the realm of clinical trials and regulatory drug approvals. Exon skipping to therapeutically restore the frame of an out-of-frame dystrophin mutation has taken center stage in drug development for DMD. The research reviewed here focuses on the clinical development of exon skipping for the treatment of DMD. In addition to the generation of clinical treatments that are being used for patient care, this research sets the stage for future therapeutic development with a focus on increasing efficacy while providing safety and addressing the multi-systemic aspects of DMD.
Collapse
Affiliation(s)
- Shin'ichi Takeda
- Honorary Director General, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Japan
| | - Paula R Clemens
- Professor and Vice Chair of VA Affairs, Department of Neurology, University of Pittsburgh School of Medicine, Division Chief, Neurology, Medical Service Line, VA Pittsburgh Healthcare System, Pittsburgh, PA USA
| | - Eric P Hoffman
- Professor, Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University - State University of New York, Binghamton, NY USA
| |
Collapse
|
30
|
Stephenson AA, Flanigan KM. Gene editing and modulation for Duchenne muscular dystrophy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 182:225-255. [PMID: 34175043 DOI: 10.1016/bs.pmbts.2021.01.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease caused by loss of dystrophin protein, encoded by the DMD gene. DMD manifests early in childhood as difficulty walking, progresses to loss of ambulation by the teens, and leads to death in early adulthood. Adeno-associated virus-vectorized gene therapies to restore dystrophin protein expression using gene replacement or antisense oligonucleotide-mediated pre-mRNA splicing modulation have emerged, making great strides in uncovering barriers to gene therapies for DMD and other genetic diseases. While this first-generation of DMD therapies are being evaluated in ongoing clinical trials, uncertainties regarding durability and therapeutic efficacy prompted the development of new experimental therapies for DMD that take advantage of somatic cell gene editing. These experimental therapies continue to advance toward clinic trials, but questions remain unanswered regarding safety and translatable efficacy. Here we review the advancements toward treatment of DMD using gene editing and modulation therapies, with an emphasis on those nearest to clinical applications.
Collapse
Affiliation(s)
- Anthony A Stephenson
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States
| | - Kevin M Flanigan
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, United States; Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH, United States; Department of Neurology, College of Medicine, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
31
|
Jackson JL, Korth CX, Leslie CE, Cotto J, Mah ML, Hor K, Cripe L, Al-Zaidy S, Camino EM, Church K, Lehman KJ, Shay V, Mendell JR. Health-Related Quality of Life and Emotional Distress Among Mothers of Sons With Muscular Dystrophy as Compared to Sex- and Age Group-Matched Controls. J Child Neurol 2021; 36:177-185. [PMID: 33034535 PMCID: PMC7854939 DOI: 10.1177/0883073820962927] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The health-related quality of life and emotional distress among mothers of sons with Duchenne or Becker muscular dystrophies (n = 82) were compared to sex- and age group-matched controls (n = 26). Participants self-reported health-related quality of life for themselves and their son(s), emotional distress, and mood/anxiety-related medication. Mothers reported poorer health-related quality of life across all domains of their health-related quality of life, as well as higher levels of emotional distress. Clinically elevated symptoms of anxiety were reported by 39% of mothers. Mothers' report of poorer health-related quality of life for their son(s) was a significant predictor of worse health-related quality of life and emotional distress for themselves across most domains. Additionally, older age of mothers predicted greater energy/less fatigue and lower levels of anxiety. Results highlight the need for screening emotional distress among mothers, as well as consideration for accessible interventions to improve the psychosocial functioning among these families.
Collapse
Affiliation(s)
- Jamie L. Jackson
- Center for Biobehavioral Health, The Abigail Wexner Research Institute at Nationwide Children’s Hospital,Department of Pediatrics, The Ohio State University
| | - Christina X. Korth
- Center for Biobehavioral Health, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Carine E. Leslie
- Center for Biobehavioral Health, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Jennifer Cotto
- Center for Biobehavioral Health, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - May Ling Mah
- The Heart Center, Nationwide Children’s Hospital
| | - Kan Hor
- The Heart Center, Nationwide Children’s Hospital
| | - Linda Cripe
- The Heart Center, Nationwide Children’s Hospital
| | | | - Eric M. Camino
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Kathleen Church
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | - Kelly J. Lehman
- Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| | | | - Jerry R. Mendell
- Department of Pediatrics, The Ohio State University,Center for Gene Therapy, The Abigail Wexner Research Institute at Nationwide Children’s Hospital
| |
Collapse
|
32
|
The lncRNA 44s2 Study Applicability to the Design of 45-55 Exon Skipping Therapeutic Strategy for DMD. Biomedicines 2021; 9:biomedicines9020219. [PMID: 33672764 PMCID: PMC7924625 DOI: 10.3390/biomedicines9020219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/10/2021] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
In skeletal muscle, long noncoding RNAs (lncRNAs) are involved in dystrophin protein stabilization but also in the regulation of myocytes proliferation and differentiation. Hence, they could represent promising therapeutic targets and/or biomarkers for Duchenne and Becker muscular dystrophy (DMD/BMD). DMD and BMD are X-linked myopathies characterized by a progressive muscular dystrophy with or without dilatative cardiomyopathy. Two-thirds of DMD gene mutations are represented by deletions, and 63% of patients carrying DMD deletions are eligible for 45 to 55 multi-exons skipping (MES), becoming BMD patients (BMDΔ45-55). We analyzed the genomic lncRNA presence in 38 BMDΔ45-55 patients and characterized the lncRNA localized in introns 44 and 55 of the DMD gene. We highlighted that all four lncRNA are differentially expressed during myogenesis in immortalized and primary human myoblasts. In addition, the lncRNA44s2 was pointed out as a possible accelerator of differentiation. Interestingly, lncRNA44s expression was associated with a favorable clinical phenotype. These findings suggest that lncRNA44s2 could be involved in muscle differentiation process and become a potential disease progression biomarker. Based on these results, we support MES45-55 therapy and propose that the design of the CRISPR/Cas9 MES45-55 assay consider the lncRNA sequences bordering the exonic 45 to 55 deletion.
Collapse
|
33
|
Jones L, Naidoo M, Machado LR, Anthony K. The Duchenne muscular dystrophy gene and cancer. Cell Oncol (Dordr) 2021; 44:19-32. [PMID: 33188621 PMCID: PMC7906933 DOI: 10.1007/s13402-020-00572-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Mutation of the Duchenne muscular dystrophy (DMD) gene causes Duchenne and Becker muscular dystrophy, degenerative neuromuscular disorders that primarily affect voluntary muscles. However, increasing evidence implicates DMD in the development of all major cancer types. DMD is a large gene with 79 exons that codes for the essential muscle protein dystrophin. Alternative promotor usage drives the production of several additional dystrophin protein products with roles that extend beyond skeletal muscle. The importance and function(s) of these gene products outside of muscle are not well understood. CONCLUSIONS We highlight a clear role for DMD in the pathogenesis of several cancers, including sarcomas, leukaemia's, lymphomas, nervous system tumours, melanomas and various carcinomas. We note that the normal balance of DMD gene products is often disrupted in cancer. The short dystrophin protein Dp71 is, for example, typically maintained in cancer whilst the full-length Dp427 gene product, a likely tumour suppressor, is frequently inactivated in cancer due to a recurrent loss of 5' exons. Therefore, the ratio of short and long gene products may be important in tumorigenesis. In this review, we summarise the tumours in which DMD is implicated and provide a hypothesis for possible mechanisms of tumorigenesis, although the question of cause or effect may remain. We hope to stimulate further study into the potential role of DMD gene products in cancer and the development of novel therapeutics that target DMD.
Collapse
Affiliation(s)
- Leanne Jones
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
| | - Michael Naidoo
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
| | - Lee R Machado
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
- Department of Genetics and Genome Science, University of Leicester, LE1 7RH, Leicester, UK
| | - Karen Anthony
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK.
| |
Collapse
|
34
|
Brandsema JF. Evaluating a Stone of Hope: ICER's 2019 Review of Treatments for Duchenne Muscular Dystrophy. J Manag Care Spec Pharm 2021; 26:366-368. [PMID: 32223600 PMCID: PMC10390954 DOI: 10.18553/jmcp.2020.26.4.366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
DISCLOSURES No funding contributed to the writing of this commentary. Brandsema reports consulting for Alexion, Audentes, AveXis, Biogen, Cytokinetics, PTC Therapeutics, Sarepta, and WaVe and has received research funding as a site investigator from Alexion, AveXis, Biogen, CSL Behring, Cytokinetics, Fibrogen, Pfizer, PTC Therapeutics, Sarepta, Summit, and WaVe.
Collapse
|
35
|
Scaglioni D, Catapano F, Ellis M, Torelli S, Chambers D, Feng L, Beck M, Sewry C, Monforte M, Harriman S, Koenig E, Malhotra J, Popplewell L, Guglieri M, Straub V, Mercuri E, Servais L, Phadke R, Morgan J, Muntoni F. The administration of antisense oligonucleotide golodirsen reduces pathological regeneration in patients with Duchenne muscular dystrophy. Acta Neuropathol Commun 2021; 9:7. [PMID: 33407808 PMCID: PMC7789286 DOI: 10.1186/s40478-020-01106-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 12/13/2020] [Indexed: 12/17/2022] Open
Abstract
During the last decade, multiple clinical trials for Duchenne muscular dystrophy (DMD) have focused on the induction of dystrophin expression using different strategies. Many of these trials have reported a clear increase in dystrophin protein following treatment. However, the low levels of the induced dystrophin protein have raised questions on its functionality. In our present study, using an unbiased, high-throughput digital image analysis platform, we assessed markers of regeneration and levels of dystrophin associated protein via immunofluorescent analysis of whole muscle sections in 25 DMD boys who received 48-weeks treatment with exon 53 skipping morpholino antisense oligonucleotide (PMO) golodirsen. We demonstrate that the de novo dystrophin induced by exon skipping with PMO golodirsen is capable of conferring a histological benefit in treated patients with an increase in dystrophin associated proteins at the dystrophin positive regions of the sarcolemma in post-treatment biopsies. Although 48 weeks treatment with golodirsen did not result in a significant change in the levels of fetal/developmental myosins for the entire cohort, there was a significant negative correlation between the amount of dystrophin and levels of regeneration observed in different biopsy samples. Our results provide, for the first time, evidence of functionality of induced dystrophin following successful therapeutic intervention in the human.
Collapse
|
36
|
Zheng J, Xu X, Zhang X, Wang X, Shu J, Cai C. Variants of CAPN3 cause limb-girdle muscular dystrophy type 2A in two Chinese families. Exp Ther Med 2020; 21:104. [PMID: 33335567 PMCID: PMC7739812 DOI: 10.3892/etm.2020.9536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 10/26/2020] [Indexed: 11/11/2022] Open
Abstract
Limb-girdle muscular dystrophies (LGMDs) are a group of neuromuscular diseases that are characterized by progressive muscle weakness. LGMD type 2A (LGMD2A), caused by variants in the calpain-3 (CAPN3) gene, is the most prevalent type. The present study aimed to analyze pathogenic CAPN3 gene variants in two pedigrees affected by LGMD2A. Each family contains three patients who are siblings and sought genetic counseling. Genomic DNA was extracted from the peripheral blood samples collected from the probands and family members and whole-exome sequencing (WES) was used to detect the pathogenic genes in the probands. Suspected variants were subsequently validated by Sanger sequencing. In family 1, WES revealed that the proband carried the compound heterogeneous variants c.1194-9A>G and c.1437C>T (p.Ser479=) in CAPN3 (NM_000070.2). In family 2, WES identified that the proband carried the compound heterogeneous variants c.632+4A>G and c.1468C>T (p.Arg490Trp) in CAPN3 (NM_000070.2). In conclusion, the present study indicated that the compound heterogeneous variants of the CAPN3 gene were most likely responsible for LGMD2A in the two Chinese families.
Collapse
Affiliation(s)
- Jie Zheng
- Graduate College, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Xiaowei Xu
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, P.R. China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, P.R. China
| | - Xinjie Zhang
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, P.R. China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, P.R. China
| | - Xuetao Wang
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, P.R. China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, P.R. China
| | - Jianbo Shu
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, P.R. China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, P.R. China
| | - Chunquan Cai
- Tianjin Pediatric Research Institute, Tianjin Children's Hospital (Children's Hospital of Tianjin University), Tianjin 300134, P.R. China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin 300134, P.R. China.,Department of Neurosurgery, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| |
Collapse
|
37
|
Lim KRQ, Nguyen Q, Yokota T. Genotype-Phenotype Correlations in Duchenne and Becker Muscular Dystrophy Patients from the Canadian Neuromuscular Disease Registry. J Pers Med 2020; 10:E241. [PMID: 33238405 PMCID: PMC7712074 DOI: 10.3390/jpm10040241] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/18/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disorder generally caused by out-of-frame mutations in the DMD gene. In contrast, in-frame mutations usually give rise to the milder Becker muscular dystrophy (BMD). However, this reading frame rule does not always hold true. Therefore, an understanding of the relationships between genotype and phenotype is important for informing diagnosis and disease management, as well as the development of genetic therapies. Here, we evaluated genotype-phenotype correlations in DMD and BMD patients enrolled in the Canadian Neuromuscular Disease Registry from 2012 to 2019. Data from 342 DMD and 60 BMD patients with genetic test results were analyzed. The majority of patients had deletions (71%), followed by small mutations (17%) and duplications (10%); 2% had negative results. Two deletion hotspots were identified, exons 3-20 and exons 45-55, harboring 86% of deletions. Exceptions to the reading frame rule were found in 13% of patients with deletions. Surprisingly, C-terminal domain mutations were associated with decreased wheelchair use and increased forced vital capacity. Dp116 and Dp71 mutations were also linked with decreased wheelchair use, while Dp140 mutations significantly predicted cardiomyopathy. Finally, we found that 12.3% and 7% of DMD patients in the registry could be treated with FDA-approved exon 51- and 53-skipping therapies, respectively.
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.); (Q.N.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G2H7, Canada
| |
Collapse
|
38
|
Meng J, Counsell J, Morgan JE. Effects of Mini-Dystrophin on Dystrophin-Deficient, Human Skeletal Muscle-Derived Cells. Int J Mol Sci 2020; 21:E7168. [PMID: 32998454 PMCID: PMC7582244 DOI: 10.3390/ijms21197168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We are developing a novel therapy for Duchenne muscular dystrophy (DMD), involving the transplantation of autologous, skeletal muscle-derived stem cells that have been genetically corrected to express dystrophin. Dystrophin is normally expressed in activated satellite cells and in differentiated muscle fibres. However, in past preclinical validation studies, dystrophin transgenes have generally been driven by constitutive promoters that would be active at every stage of the myogenic differentiation process, including in proliferating muscle stem cells. It is not known whether artificial dystrophin expression would affect the properties of these cells. AIMS Our aims are to determine if mini-dystrophin expression affects the proliferation or myogenic differentiation of DMD skeletal muscle-derived cells. METHODS Skeletal muscle-derived cells from a DMD patient were transduced with lentivirus coding for mini-dystrophins (R3-R13 spectrin-like repeats (ΔR3R13) or hinge2 to spectrin-like repeats R23 (ΔH2R23)) with EGFP (enhanced green fluorescence protein) fused to the C-terminus, driven by a constitutive promoter, spleen focus-forming virus (SFFV). Transduced cells were purified on the basis of GFP expression. Their proliferation and myogenic differentiation were quantified by ethynyl deoxyuridine (EdU) incorporation and fusion index. Furthermore, dystrophin small interfering ribonucleic acids (siRNAs) were transfected to the cells to reverse the effects of the mini-dystrophin. Finally, a phospho-mitogen-activated protein kinase (MAPK) array assay was performed to investigate signalling pathway changes caused by dystrophin expression. RESULTS Cell proliferation was not affected in cells transduced with ΔR3R13, but was significantly increased in cells transduced with ΔH2R23. The fusion index of myotubes derived from both ΔR3R13- and ΔH2R23 -expressing cells was significantly compromised in comparison to myotubes derived from non-transduced cells. Dystrophin siRNA transfection restored the differentiation of ΔH2R23-expressing cells. The Erk1/2- signalling pathway is altered in cells transduced with mini-dystrophin constructs. CONCLUSIONS Ectopic expression of dystrophin in cultured human skeletal muscle-derived cells may affect their proliferation and differentiation capacity. Caution should be taken when considering genetic correction of autologous stem cells to express dystrophin driven by a constitutive promoter.
Collapse
MESH Headings
- Cell Differentiation
- Cell Engineering/methods
- Cell Proliferation
- Dystrophin/antagonists & inhibitors
- Dystrophin/genetics
- Dystrophin/metabolism
- Gene Expression Regulation
- Genes, Reporter
- Genetic Vectors/chemistry
- Genetic Vectors/metabolism
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Humans
- Lentivirus/genetics
- Lentivirus/metabolism
- MAP Kinase Signaling System
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Plasmids/chemistry
- Plasmids/metabolism
- Primary Cell Culture
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Spectrin/genetics
- Spectrin/metabolism
- Transduction, Genetic
- Transgenes
Collapse
Affiliation(s)
- Jinhong Meng
- Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (J.M.); (J.C.)
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK
| | - John Counsell
- Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (J.M.); (J.C.)
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK
| | - Jennifer E. Morgan
- Dubowitz Neuromuscular Centre, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK; (J.M.); (J.C.)
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 1EH, UK
| |
Collapse
|
39
|
Teramoto N, Sugihara H, Yamanouchi K, Nakamura K, Kimura K, Okano T, Shiga T, Shirakawa T, Matsuo M, Nagata T, Daimon M, Matsuwaki T, Nishihara M. Pathological evaluation of rats carrying in-frame mutations in the dystrophin gene: a new model of Becker muscular dystrophy. Dis Model Mech 2020; 13:dmm044701. [PMID: 32859695 PMCID: PMC7541341 DOI: 10.1242/dmm.044701] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/18/2020] [Indexed: 01/10/2023] Open
Abstract
Dystrophin, encoded by the DMD gene on the X chromosome, stabilizes the sarcolemma by linking the actin cytoskeleton with the dystrophin-glycoprotein complex (DGC). In-frame mutations in DMD cause a milder form of X-linked muscular dystrophy, called Becker muscular dystrophy (BMD), characterized by the reduced expression of truncated dystrophin. So far, no animal model with in-frame mutations in Dmd has been established. As a result, the effect of in-frame mutations on the dystrophin expression profile and disease progression of BMD remains unclear. In this study, we established a novel rat model carrying in-frame Dmd gene mutations (IF rats) and evaluated the pathology. We found that IF rats exhibited reduced expression of truncated dystrophin in a proteasome-independent manner. This abnormal dystrophin expression caused dystrophic changes in muscle tissues but did not lead to functional deficiency. We also found that the expression of additional dystrophin named dpX, which forms the DGC in the sarcolemma, was associated with the appearance of truncated dystrophin. In conclusion, the outcomes of this study contribute to the further understanding of BMD pathology and help elucidate the efficiency of dystrophin recovery treatments in Duchenne muscular dystrophy, a more severe form of X-linked muscular dystrophy.
Collapse
Affiliation(s)
- Naomi Teramoto
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Hidetoshi Sugihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Katsuyuki Nakamura
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Koichi Kimura
- Department of General Medicine, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, 108-8639, Japan
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Tomoko Okano
- Department of Laboratory Medicine, The University of Tokyo Hospital, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takanori Shiga
- Department of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Taku Shirakawa
- Research Center for Locomotion Biology, Kobe Gakuin University, Nishi, Kobe, 651-2180, Japan
- KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Nishi, Kobe, 651-2180, Japan
| | - Masafumi Matsuo
- Research Center for Locomotion Biology, Kobe Gakuin University, Nishi, Kobe, 651-2180, Japan
- KNC Department of Nucleic Acid Drug Discovery, Faculty of Rehabilitation, Kobe Gakuin University, Nishi, Kobe, 651-2180, Japan
| | - Tetsuya Nagata
- Department of Neurology and Neurological Science, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Masao Daimon
- Department of Laboratory Medicine, The University of Tokyo Hospital, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Matsuwaki
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, 113-8657, Japan
| |
Collapse
|
40
|
Torella A, Zanobio M, Zeuli R, del Vecchio Blanco F, Savarese M, Giugliano T, Garofalo A, Piluso G, Politano L, Nigro V. The position of nonsense mutations can predict the phenotype severity: A survey on the DMD gene. PLoS One 2020; 15:e0237803. [PMID: 32813700 PMCID: PMC7437896 DOI: 10.1371/journal.pone.0237803] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/03/2020] [Indexed: 12/23/2022] Open
Abstract
A nonsense mutation adds a premature stop signal that hinders any further translation of a protein-coding gene, usually resulting in a null allele. To investigate the possible exceptions, we used the DMD gene as an ideal model. First, because dystrophin absence causes Duchenne muscular dystrophy (DMD), while its reduction causes Becker muscular dystrophy (BMD). Second, the DMD gene is X-linked and there is no second allele that can interfere in males. Third, databases are accumulating reports on many mutations and phenotypic data. Finally, because DMD mutations may have important therapeutic implications. For our study, we analyzed large databases (LOVD, HGMD and ClinVar) and literature and revised critically all data, together with data from our internal patients. We totally collected 2593 patients. Positioning these mutations along the dystrophin transcript, we observed a nonrandom distribution of BMD-associated mutations within selected exons and concluded that the position can be predictive of the phenotype. Nonsense mutations always cause DMD when occurring at any point in fifty-one exons. In the remaining exons, we found milder BMD cases due to early 5’ nonsense mutations, if reinitiation can occur, or due to late 3’ nonsense when the shortened product retains functionality. In the central part of the gene, all mutations in some in-frame exons, such as in exons 25, 31, 37 and 38 cause BMD, while mutations in exons 30, 32, 34 and 36 cause DMD. This may have important implication in predicting the natural history and the efficacy of therapeutic use of drug-stimulated translational readthrough of premature termination codons, also considering the action of internal natural rescuers. More in general, our survey confirm that a nonsense mutation should be not necessarily classified as a null allele and this should be considered in genetic counselling.
Collapse
Affiliation(s)
- Annalaura Torella
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Mariateresa Zanobio
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Roberta Zeuli
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | | | - Marco Savarese
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
- Folkhälsan Research Center, Medicum, University of Helsinki, Helsinki, Finland
| | - Teresa Giugliano
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Arcomaria Garofalo
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Giulio Piluso
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Luisa Politano
- Dipartimento di Medicina Sperimentale, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Vincenzo Nigro
- Dipartimento di Medicina di Precisione, Università degli Studi della Campania “Luigi Vanvitelli”, Napoli, Italy
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- * E-mail:
| |
Collapse
|
41
|
Wonkam‐Tingang E, Nguefack S, Esterhuizen AI, Chelo D, Wonkam A. DMD-related muscular dystrophy in Cameroon: Clinical and genetic profiles. Mol Genet Genomic Med 2020; 8:e1362. [PMID: 32543101 PMCID: PMC7434738 DOI: 10.1002/mgg3.1362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Most of the previous studies on Duchenne Muscular Dystrophy (DMD) were conducted in Caucasian, Asian, and Arab populations. Therefore, little is known about the features of this disease in Africans. In this study, we aimed to determine the clinical characteristics of DMD, and the common mutations associated with this condition in a group of Cameroonian patients. METHODS We recruited DMD patients and performed a general physical examination on each of them. Multiplex ligand-dependant probe amplification was carried out to investigate exon deletions and duplications in the DMD gene (OMIM: 300377) of patients and their mothers. RESULTS A total of 17 male patients from 14 families were recruited, aged 14 ± 5.1 (8-23) years. The mean age at onset of symptoms was 4.6 ± 1.5 years, and the mean age at diagnosis was 12.1 ± 5.2 years. Proximal muscle weakness was noted in all patients and calf hypertrophy in the large majority of them (88.2%; 15/17). Flexion contractures were particularly frequent on the ankle (85.7%; 12/14). Wasting of shoulder girdle and thigh muscles was present in 50% (6/12) and 46.2% (6/13) of patients, respectively. No patient presented with hearing impairment. Deletions in DMD gene (OMIM: 300377) occurred in 45.5% of patients (5/11), while duplications were observed in 27.3% (3/11). Both mutation types were clustered between exons 45 and 50, and the proportion of de novo mutation was estimated at 18.2% (2/11). CONCLUSION Despite the first symptoms of DMD occurring in infancy, the diagnosis is frequently made later in adolescence, indicating an underestimation of the number of cases of DMD in Cameroon. Future screening of deletions and duplications in patients from Cameroon should focus on the distal part of the gene.
Collapse
Affiliation(s)
- Edmond Wonkam‐Tingang
- Division of Human GeneticsDepartment of PathologyUniversity of Cape TownCape TownSouth Africa
| | - Séraphin Nguefack
- Department of PaediatricsFaculty of Medicine and Biomedical SciencesUniversity of Yaoundé IYaoundéCameroon
- Paediatrics UnitDivision of Paediatric NeurologyGynaeco‐Obstetric and Paediatric HospitalYaoundéCameroon
| | - Alina I. Esterhuizen
- Division of Human GeneticsDepartment of PathologyUniversity of Cape TownCape TownSouth Africa
- National Health Laboratory ServiceGroote Schuur HospitalCape TownSouth Africa
| | - David Chelo
- Department of PaediatricsFaculty of Medicine and Biomedical SciencesUniversity of Yaoundé IYaoundéCameroon
- Division of Paediatric Cardiology, Mother and Child HospitalYaoundéCameroon
| | - Ambroise Wonkam
- Division of Human GeneticsDepartment of PathologyUniversity of Cape TownCape TownSouth Africa
- Department of MedicineUniversity of Cape TownCape TownSouth Africa
| |
Collapse
|
42
|
Clemens PR, Niizawa G, Feng J, Florence J, DʼAlessandro AS, Morgenroth LP, Gorni K, Guglieri M, Connolly A, Wicklund M, Bertorini T, Mah JK, Thangarajh M, Smith E, Kuntz N, McDonald CM, Henricson EK, Upadhyayula S, Byrne B, Manousakis G, Harper A, Bravver E, Iannaccone S, Spurney C, Cnaan A, Gordish-Dressman H. The CINRG Becker Natural History Study: Baseline characteristics. Muscle Nerve 2020; 62:369-376. [PMID: 32564389 DOI: 10.1002/mus.27011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 01/16/2023]
Abstract
We performed an observational, natural history study of males with in-frame dystrophin gene deletions causing Becker muscular dystrophy (BMD). A prospective natural history study collected longitudinal medical, strength, and timed function assessments. Eighty-three participants with genetically confirmed BMD were enrolled (age range 5.6-75.4 years). Lower extremity function and the percentage of participants who retained ambulation declined across the age span. The largest single group of participants had in-frame deletions that corresponded to an out-of-frame deletion treated with an exon 45 skip to restore the reading frame. This group of 54 participants showed similarities in baseline motor functional assessments when compared to the group of all others in the study. A prospective natural history cohort with in-frame dystrophin gene deletions offers the potential to contribute to clinical trial readiness for BMD and to analyze therapeutic benefit of exon skipping for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Paula R Clemens
- Department of Veterans Affairs Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Gabriela Niizawa
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jia Feng
- Children's National Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | - Jean K Mah
- Alberta Children's Hospital, Calgary, Alberta, Canada
| | | | - Edward Smith
- Duke University Medical Center, Durham, North Carolina, USA
| | - Nancy Kuntz
- Lurie Children's Hospital, Chicago, Illinois, USA
| | | | | | | | - Barry Byrne
- University of Florida, Gainesville, Florida, USA
| | | | - Amy Harper
- Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Elena Bravver
- Carolinas Medical Center, Charlotte, North Carolina, USA
| | - Susan Iannaccone
- University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | | | - Avital Cnaan
- Children's National Medical Center, Washington, DC, USA
| | | | | |
Collapse
|
43
|
Schüssler SC, Gerhalter T, Abicht A, Müller-Felber W, Nagel AM, Trollmann R. Rare intronic mutation between Exon 62 and 63 (c.9225-285A>G) of the dystrophin gene associated with atypical BMD phenotype. Neuromuscul Disord 2020; 30:680-684. [PMID: 32669210 DOI: 10.1016/j.nmd.2020.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Dystrophinopathies are predominantly caused by deletions, duplications and point mutations in the coding regions of the dystrophin gene with less than 1% of all pathogenic mutations identified within intronic sequences. We describe a 17-year-old male with a Becker muscular dystrophy diagnosis and mental disability due to an intron mutation that led to aberrant splicing and formation of an additional exon. Histopathological analysis of muscle tissue revealed signs of muscular dystrophy and reduced signal for dystrophin, alpha-sarcoglycan, and alpha-dystroglycan. Multiplex ligation-dependent probe amplification screening and total sequencing of the dystrophin gene did not identify a mutation in the coding regions. However, next generation sequencing revealed an intron mutation between exons 62 and 63 of the dystrophin gene known for pseudoexon formation and disruption of the reading frame. We report a functional consequence of this mutation as an increased intracellular-weighted sodium signal (assessed by 23Na-magnetic resonance imaging) in leg muscles.
Collapse
Affiliation(s)
- S C Schüssler
- Department of Pediatrics, Division of Neuropediatrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen (FAU), Loschgestr. 15, 91054 Erlangen, Germany
| | - T Gerhalter
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen (FAU), Erlangen, Germany
| | - A Abicht
- Medical Center of Human Genetics, Munich, Germany
| | - W Müller-Felber
- Department of Pediatric Neurology and Developmental Medicine, Dr. v. Hauner Children's Hospital, Ludwig-Maximilian-Universität Munich, Munich, Germany
| | - A M Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen (FAU), Erlangen, Germany
| | - R Trollmann
- Department of Pediatrics, Division of Neuropediatrics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen (FAU), Loschgestr. 15, 91054 Erlangen, Germany.
| |
Collapse
|
44
|
van Westering TLE, Lomonosova Y, Coenen-Stass AML, Betts CA, Bhomra A, Hulsker M, Clark LE, McClorey G, Aartsma-Rus A, van Putten M, Wood MJA, Roberts TC. Uniform sarcolemmal dystrophin expression is required to prevent extracellular microRNA release and improve dystrophic pathology. J Cachexia Sarcopenia Muscle 2020; 11:578-593. [PMID: 31849191 PMCID: PMC7113513 DOI: 10.1002/jcsm.12506] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disorder caused by genetic loss of dystrophin protein. Extracellular microRNAs (ex-miRNAs) are putative, minimally invasive biomarkers of DMD. Specific ex-miRNAs (e.g. miR-1, miR-133a, miR-206, and miR-483) are highly up-regulated in the serum of DMD patients and dystrophic animal models and are restored to wild-type levels following exon skipping-mediated dystrophin rescue in mdx mice. As such, ex-miRNAs are promising pharmacodynamic biomarkers of exon skipping efficacy. Here, we aimed to determine the degree to which ex-miRNA levels reflect the underlying level of dystrophin protein expression in dystrophic muscle. METHODS Candidate ex-miRNA biomarker levels were investigated in mdx mice in which dystrophin was restored with peptide-PMO (PPMO) exon skipping conjugates and in mdx-XistΔhs mice that express variable amounts of dystrophin from birth as a consequence of skewed X-chromosome inactivation. miRNA profiling was performed in mdx-XistΔhs mice using the FirePlex methodology and key results validated by small RNA TaqMan RT-qPCR. The muscles from each animal model were further characterized by dystrophin western blot and immunofluorescence staining. RESULTS The restoration of ex-myomiR abundance observed following PPMO treatment was not recapitulated in the high dystrophin-expressing mdx-XistΔhs group, despite these animals expressing similar amounts of total dystrophin protein (~37% of wild-type levels). Instead, ex-miRNAs were present at high levels in mdx-XistΔhs mice regardless of dystrophin expression. PPMO-treated muscles exhibited a uniform pattern of dystrophin localization and were devoid of regenerating fibres, whereas mdx-XistΔhs muscles showed non-homogeneous dystrophin staining and sporadic regenerating foci. CONCLUSIONS Uniform dystrophin expression is required to prevent ex-miRNA release, stabilize myofiber turnover, and attenuate pathology in dystrophic muscle.
Collapse
Affiliation(s)
- Tirsa L E van Westering
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Yulia Lomonosova
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Margriet Hulsker
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lucy E Clark
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK.,Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA, USA
| |
Collapse
|
45
|
Naidoo M, Anthony K. Dystrophin Dp71 and the Neuropathophysiology of Duchenne Muscular Dystrophy. Mol Neurobiol 2020; 57:1748-1767. [PMID: 31836945 PMCID: PMC7060961 DOI: 10.1007/s12035-019-01845-w] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by frameshift mutations in the DMD gene that prevent the body-wide translation of its protein product, dystrophin. Besides a severe muscle phenotype, cognitive impairment and neuropsychiatric symptoms are prevalent. Dystrophin protein 71 (Dp71) is the major DMD gene product expressed in the brain and mutations affecting its expression are associated with the DMD neuropsychiatric syndrome. As with dystrophin in muscle, Dp71 localises to dystrophin-associated protein complexes in the brain. However, unlike in skeletal muscle; in the brain, Dp71 is alternatively spliced to produce many isoforms with differential subcellular localisations and diverse cellular functions. These include neuronal differentiation, adhesion, cell division and excitatory synapse organisation as well as nuclear functions such as nuclear scaffolding and DNA repair. In this review, we first describe brain involvement in DMD and the abnormalities observed in the DMD brain. We then review the gene expression, RNA processing and functions of Dp71. We review genotype-phenotype correlations and discuss emerging cellular/tissue evidence for the involvement of Dp71 in the neuropathophysiology of DMD. The literature suggests changes observed in the DMD brain are neurodevelopmental in origin and that their risk and severity is associated with a cumulative loss of distal DMD gene products such as Dp71. The high risk of neuropsychiatric syndromes in Duchenne patients warrants early intervention to achieve the best possible quality of life. Unravelling the function and pathophysiological significance of dystrophin in the brain has become a high research priority to inform the development of brain-targeting treatments for Duchenne.
Collapse
Affiliation(s)
- Michael Naidoo
- Centre for Physical Activity and Life Sciences, Faculty of Arts, Science and Technology, University of Northampton, University Drive, Northampton, Northamptonshire, NN1 5PH, UK
| | - Karen Anthony
- Centre for Physical Activity and Life Sciences, Faculty of Arts, Science and Technology, University of Northampton, University Drive, Northampton, Northamptonshire, NN1 5PH, UK.
| |
Collapse
|
46
|
Le Guen YT, Le Gall T, Midoux P, Guégan P, Braun S, Montier T. Gene transfer to skeletal muscle using hydrodynamic limb vein injection: current applications, hurdles and possible optimizations. J Gene Med 2020; 22:e3150. [PMID: 31785130 DOI: 10.1002/jgm.3150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 11/06/2022] Open
Abstract
Hydrodynamic limb vein injection is an in vivo locoregional gene delivery method. It consists of administrating a large volume of solution containing nucleic acid constructs in a limb with both blood inflow and outflow temporarily blocked using a tourniquet. The fast, high pressure delivery allows the musculature of the whole limb to be reached. The skeletal muscle is a tissue of choice for a variety of gene transfer applications, including gene therapy for Duchenne muscular dystrophy or other myopathies, as well as for the production of antibodies or other proteins with broad therapeutic effects. Hydrodynamic limb vein delivery has been evaluated with success in a large range of animal models. It has also proven to be safe and well-tolerated in muscular dystrophy patients, thus supporting its translation to the clinic. However, some possible limitations may occur at different steps of the delivery process. Here, we have highlighted the interests, bottlenecks and potential improvements that could further optimize non-viral gene transfer following hydrodynamic limb vein injection.
Collapse
Affiliation(s)
| | - Tony Le Gall
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d'Orléans, France
| | - Philippe Guégan
- Laboratoire de chimie des polymères, Sorbonne Université, CNRS UMR 8232, UPMC Paris 06, F-75005, Paris, France
| | - Serge Braun
- AFM Telethon, 1 rue de l'Internationale, BP59, 91002 Evry, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France.,Service de Génétique Médicale et Biologie de la Reproduction, Centre de référence des maladies rares 'Maladies neuromusculaires', CHRU de Brest, F-29200, Brest, France
| |
Collapse
|
47
|
Aartsma-Rus A, Morgan J, Lonkar P, Neubert H, Owens J, Binks M, Montolio M, Phadke R, Datson N, Van Deutekom J, Morris GE, Rao VA, Hoffman EP, Muntoni F, Arechavala-Gomeza V. Report of a TREAT-NMD/World Duchenne Organisation Meeting on Dystrophin Quantification Methodology. J Neuromuscul Dis 2020; 6:147-159. [PMID: 30614809 PMCID: PMC6398559 DOI: 10.3233/jnd-180357] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Representatives of academia, patient organisations, industry and the United States Food and Drug Administration attended a workshop on dystrophin quantification methodology. The aims of the workshop were to provide an overview of methods used to quantify dystrophin levels in human skeletal muscle and their applicability to clinical trial samples, outline the gaps with regards to validating the methods for robust clinical applications prior to regulatory agency review, and to align future efforts towards further optimizing these methods. The workshop facilitated a constructive but also critical discussion on the potential and limitations of techniques currently used in the field of translational research (western blot and immunofluorescence analysis) and emerging techniques (mass spectrometry and capillary western immunoassay). Notably, all participants reported variation in dystrophin levels between muscle biopsies from different healthy individuals and agreed on the need for a common reference sample.
Collapse
Affiliation(s)
| | - Jennifer Morgan
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Hendrik Neubert
- Pfizer Inc, BioMedicine Design 1 Burtt Road, Andover, MA, USA
| | - Jane Owens
- Pfizer Inc, Rare Disease Research Unit, 610 Main Street, Cambridge, MA, USA
| | - Michael Binks
- Pfizer Inc, Rare Disease Research Unit, 610 Main Street, Cambridge, MA, USA
| | - Marisol Montolio
- Department of Cell Biology, Fisiology and Immunology, Faculty of Biology, University of Barcelona.,Duchenne Parent Project Spain, Spain
| | - Rahul Phadke
- National Hospital for Neurology and Neurosurgery, UCL Institute of Neurology, London, United Kingdom
| | | | | | - Glenn E Morris
- Wolfson Centre for Inherited Neuromuscular Disease, Keele University and RJAH Orthopaedic Hospital, Oswestry, UK
| | - V Ashutosh Rao
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, MD, USA
| | - Eric P Hoffman
- Binghamton University-SUNY, Binghamton, NY, USA and AGADA BioSciences, Halifax, Nova Scotia, Canada
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1, N 1EH, United Kingdom
| | | | | |
Collapse
|
48
|
Dietz AR, Connolly A, Dori A, Zaidman CM. Intramuscular blood flow in Duchenne and Becker Muscular Dystrophy: Quantitative power Doppler sonography relates to disease severity. Clin Neurophysiol 2019; 131:1-5. [PMID: 31751835 DOI: 10.1016/j.clinph.2019.09.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 08/02/2019] [Accepted: 09/11/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Absent or truncated dystrophin in Duchenne (DMD) and Becker (BMD) muscular dystrophies results in impaired vasodilatory pathways and exercise induced muscle ischemia. Here, we used power Doppler sonography to quantify changes in intramuscular blood flow immediately following exercise in boys with D/BMD. METHOD We quantified changes in intramuscular blood flow following exercise using power Doppler sonography in 14 boys with D/BMD and compared changes in muscle blood flow to disease severity and to historic controls. RESULT Post exercise blood flow change in the anterior forearm muscles is lower in (1) DMD (median 0.25%; range -0.47 to 2.19%) than BMD (2.46%; 2.02-3.38%, p < 0.05) and historical controls (6.59%; 2.16-12.40%, p < 0.01); (2) in non-ambulatory (0.04%; -0.47 to 0.10%) than ambulatory DMD boys (0.71%; 0.07-2.19%, p < 0.05); and (3) in muscle with higher echointensity (rs = -0.7253, p = 0.005). The tibialis anterior showed similar findings. We estimate that a single sample clinical trial would require 19 subjects to detect a doubling of blood flow to the anterior forearm after the intervention. CONCLUSION Post-exercise blood flow is reduced in D/BMD and relates to disease severity. SIGNIFICANCE Our protocol for quantifying post-exercise intramuscular blood flow is feasible for clinical trials in D/BMD.
Collapse
Affiliation(s)
- Alexander R Dietz
- Blue Sky Neurology, Englewood, CO, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne Connolly
- Department of Pediatrics, Division of Neurology, Nationwide Children's Hospital, Columbus OH, USA
| | - Amir Dori
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA; Department of Neurology, Talpiot Medical Leadership Program, Chaim Sheba Medical Center, Tel HaShomer, and Joseph Sagol Neuroscience Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Craig M Zaidman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
49
|
Boehler JF, Ricotti V, Gonzalez JP, Soustek-Kramer M, Such L, Brown KJ, Schneider JS, Morris CA. Membrane recruitment of nNOSµ in microdystrophin gene transfer to enhance durability. Neuromuscul Disord 2019; 29:735-741. [PMID: 31521486 DOI: 10.1016/j.nmd.2019.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022]
Abstract
Several gene transfer clinical trials are currently ongoing with the common aim of delivering a shortened version of dystrophin, termed a microdystrophin, for the treatment of Duchenne muscular dystrophy (DMD). However, one of the main differences between these trials is the microdystrophin protein produced following treatment. Each gene transfer product is based on different selections of dystrophin domain combinations to assemble microdystrophin transgenes that maintain functional dystrophin domains and fit within the packaging limits of an adeno-associated virus (AAV) vector. While domains involved in mechanical function, such as the actin-binding domain and β-dystroglycan binding domain, have been identified for many years and included in microdystrophin constructs, more recently the neuronal nitric oxide synthase (nNOS) domain has also been identified due to its role in enhancing nNOS membrane localization. As nNOS membrane localization has been established as an important requirement for prevention of functional ischemia in skeletal muscle, inclusion of the nNOS domain into a microdystrophin construct represents an important consideration. The aim of this mini review is to highlight what is currently known about the nNOS domain of dystrophin and to describe potential implications of this domain in a microdystrophin gene transfer clinical trial.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Valeria Ricotti
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - J Patrick Gonzalez
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | | | - Lauren Such
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Kristy J Brown
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Joel S Schneider
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Carl A Morris
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States.
| |
Collapse
|
50
|
Echigoya Y, Lim KRQ, Melo D, Bao B, Trieu N, Mizobe Y, Maruyama R, Mamchaoui K, Tanihata J, Aoki Y, Takeda S, Mouly V, Duddy W, Yokota T. Exons 45-55 Skipping Using Mutation-Tailored Cocktails of Antisense Morpholinos in the DMD Gene. Mol Ther 2019; 27:2005-2017. [PMID: 31416775 DOI: 10.1016/j.ymthe.2019.07.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/09/2019] [Accepted: 07/15/2019] [Indexed: 12/16/2022] Open
Abstract
Mutations in the dystrophin (DMD) gene and consequent loss of dystrophin cause Duchenne muscular dystrophy (DMD). A promising therapy for DMD, single-exon skipping using antisense phosphorodiamidate morpholino oligomers (PMOs), currently confronts major issues in that an antisense drug induces the production of functionally undefined dystrophin and may not be similarly efficacious among patients with different mutations. Accordingly, the applicability of this approach is limited to out-of-frame mutations. Here, using an exon-skipping efficiency predictive tool, we designed three different PMO cocktail sets for exons 45-55 skipping aiming to produce a dystrophin variant with preserved functionality as seen in milder or asymptomatic individuals with an in-frame exons 45-55 deletion. Of them, the most effective set was composed of select PMOs that each efficiently skips an assigned exon in cell-based screening. These combinational PMOs fitted to different deletions of immortalized DMD patient muscle cells significantly induced exons 45-55 skipping with removing 3, 8, or 10 exons and dystrophin restoration as represented by western blotting. In vivo skipping of the maximum 11 human DMD exons was confirmed in humanized mice. The finding indicates that our PMO set can be used to create mutation-tailored cocktails for exons 45-55 skipping and treat over 65% of DMD patients carrying out-of-frame or in-frame deletions.
Collapse
Affiliation(s)
- Yusuke Echigoya
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Laboratory of Biomedical Science, Department of Veterinary Medicine, College of Bioresource Sciences, Nihon University, Fujisawa, Kanagawa 252-0880, Japan
| | - Kenji Rowel Q Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Dyanna Melo
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Bo Bao
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Nhu Trieu
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Yoshitaka Mizobe
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Kamel Mamchaoui
- UPMC-Sorbonne Universités-University Paris 6, UPMC/INSERM UMRS974, CNRS FRE 3617, Myology Centre for Research, Paris Cedex 13 75651, France
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan; Department of Cell Physiology, The Jikei University School of Medicine, Minato, Tokyo 105-8461, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8502, Japan
| | - Vincent Mouly
- UPMC-Sorbonne Universités-University Paris 6, UPMC/INSERM UMRS974, CNRS FRE 3617, Myology Centre for Research, Paris Cedex 13 75651, France
| | - William Duddy
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry BT47 6SB, UK
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Muscular Dystrophy Canada Research Chair, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|