1
|
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
|
2
|
Gatto F, Benemei S, Piluso G, Bello L. The complex landscape of DMD mutations: moving towards personalized medicine. Front Genet 2024; 15:1360224. [PMID: 38596212 PMCID: PMC11002111 DOI: 10.3389/fgene.2024.1360224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration, with respiratory and cardiac complications, caused by mutations in the DMD gene, encoding the protein dystrophin. Various DMD mutations result in different phenotypes and disease severity. Understanding genotype/phenotype correlations is essential to optimize clinical care, as mutation-specific therapies and innovative therapeutic approaches are becoming available. Disease modifier genes, trans-active variants influencing disease severity and phenotypic expressivity, may modulate the response to therapy, and become new therapeutic targets. Uncovering more disease modifier genes via extensive genomic mapping studies offers the potential to fine-tune prognostic assessments for individuals with DMD. This review provides insights into genotype/phenotype correlations and the influence of modifier genes in DMD.
Collapse
Affiliation(s)
| | | | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Aljaberi R, Vengoechea J. Isolated cardiomyopathy in a pathogenic X-linked in frame hemizygous DMD exon 49 deletion: A rare presentation with normal creatine kinase levels and absence of musculoskeletal symptoms. Am J Med Genet A 2024; 194:e63475. [PMID: 37961025 DOI: 10.1002/ajmg.a.63475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Affiliation(s)
- Rana Aljaberi
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| | - Jaime Vengoechea
- Department of Human Genetics, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Shiba N, Yang X, Sato M, Kadota S, Suzuki Y, Agata M, Nagamine K, Izumi M, Honda Y, Koganehira T, Kobayashi H, Ichimura H, Chuma S, Nakai J, Tohyama S, Fukuda K, Miyazaki D, Nakamura A, Shiba Y. Efficacy of exon-skipping therapy for DMD cardiomyopathy with mutations in actin binding domain 1. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102060. [PMID: 38028197 PMCID: PMC10654596 DOI: 10.1016/j.omtn.2023.102060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 10/17/2023] [Indexed: 12/01/2023]
Abstract
Exon-skipping therapy is a promising treatment strategy for Duchenne muscular dystrophy (DMD), which is caused by loss-of-function mutations in the DMD gene encoding dystrophin, leading to progressive cardiomyopathy. In-frame deletion of exons 3-9 (Δ3-9), manifesting a very mild clinical phenotype, is a potential targeted reading frame for exon-skipping by targeting actin-binding domain 1 (ABD1); however, the efficacy of this approach for DMD cardiomyopathy remains uncertain. In this study, we compared three isogenic human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) expressing Δ3-9, frameshifting Δ3-7, or intact DMD. RNA sequencing revealed a resemblance in the expression patterns of mechano-transduction-related genes between Δ3-9 and wild-type samples. Furthermore, we observed similar electrophysiological properties between Δ3-9 and wild-type hiPSC-CMs; Δ3-7 hiPSC-CMs showed electrophysiological alterations with accelerated CaMKII activation. Consistently, Δ3-9 hiPSC-CMs expressed substantial internally truncated dystrophin protein, resulting in maintaining F-actin binding and desmin retention. Antisense oligonucleotides targeting exon 8 efficiently induced skipping exons 8-9 to restore functional dystrophin and electrophysiological parameters in Δ3-7 hiPSC-CMs, bringing the cell characteristics closer to those of Δ3-9 hiPSC-CMs. Collectively, exon-skipping targeting ABD1 to convert the reading frame to Δ3-9 may become a promising therapy for DMD cardiomyopathy.
Collapse
Affiliation(s)
- Naoko Shiba
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
- Department of Pediatrics, Shinshu University, Matsumoto 390-8621, Japan
| | - Xiao Yang
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Mitsuto Sato
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Shin Kadota
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
- Institute for Biomedical Sciences, Shinshu University, Matsumoto 390-8621, Japan
| | - Yota Suzuki
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Masahiro Agata
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Kohei Nagamine
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Masaki Izumi
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Yusuke Honda
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Tomoya Koganehira
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Hideki Kobayashi
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Hajime Ichimura
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
| | - Shinichiro Chuma
- Department of Regeneration Science and Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Junichi Nakai
- Graduate Schools of Dentistry, Tohoku University, Sendai 980-8575, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Daigo Miyazaki
- Department of Medicine (Neurology and Rheumatology), Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Akinori Nakamura
- Department of Clinical Research, National Hospital Organization Matsumoto Medical Center, Matsumoto 399-8701, Japan
| | - Yuji Shiba
- Department of Regenerative Science and Medicine, Shinshu University, Matsumoto 390-8621, Japan
- Institute for Biomedical Sciences, Shinshu University, Matsumoto 390-8621, Japan
| |
Collapse
|
7
|
Kita Y, Okuzaki Y, Naoe Y, Lee J, Bang U, Okawa N, Ichiki A, Jonouchi T, Sakurai H, Kojima Y, Hotta A. Dual CRISPR-Cas3 system for inducing multi-exon skipping in DMD patient-derived iPSCs. Stem Cell Reports 2023; 18:1753-1765. [PMID: 37625413 PMCID: PMC10545483 DOI: 10.1016/j.stemcr.2023.07.007] [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: 10/03/2022] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
To restore dystrophin protein in various mutation patterns of Duchenne muscular dystrophy (DMD), the multi-exon skipping (MES) approach has been investigated. However, only limited techniques are available to induce a large deletion to cover the target exons spread over several hundred kilobases. Here, we utilized the CRISPR-Cas3 system for MES induction and showed that dual crRNAs could induce a large deletion at the dystrophin exon 45-55 region (∼340 kb), which can be applied to various types of DMD patients. We developed a two-color SSA-based reporter system for Cas3 to enrich the genome-edited cell population and demonstrated that MES induction restored dystrophin protein in DMD-iPSCs with three distinct mutations. Whole-genome sequencing and distance analysis detected no significant off-target deletion near the putative crRNA binding sites. Altogether, dual CRISPR-Cas3 is a promising tool to induce a gigantic genomic deletion and restore dystrophin protein via MES induction.
Collapse
Affiliation(s)
- Yuto Kita
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuya Okuzaki
- Nagoya University Graduate School of Bioagricultural Sciences, Avian Bioscience Research Center, Furo-cho, Chikusa-ku, Nagoya, Aishi 464-8601, Japan
| | - Youichi Naoe
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Joseph Lee
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Uikyu Bang
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Natsumi Okawa
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akane Ichiki
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuya Jonouchi
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yusuke Kojima
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application, Kyoto University, 53 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Takeda-CiRA Joint Program (T-CiRA), Fujisawa, Kanagawa 251-8555, Japan.
| |
Collapse
|
8
|
Saad FA, Siciliano G, Angelini C. Advances in Dystrophinopathy Diagnosis and Therapy. Biomolecules 2023; 13:1319. [PMID: 37759719 PMCID: PMC10526396 DOI: 10.3390/biom13091319] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Dystrophinopathies are x-linked muscular disorders which emerge from mutations in the Dystrophin gene, including Duchenne and Becker muscular dystrophy, and dilated cardiomyopathy. However, Duchenne muscular dystrophy interconnects with bone loss and osteoporosis, which are exacerbated by glucocorticoids therapy. Procedures for diagnosing dystrophinopathies include creatine kinase assay, haplotype analysis, Southern blot analysis, immunological analysis, multiplex PCR, multiplex ligation-dependent probe amplification, Sanger DNA sequencing, and next generation DNA sequencing. Pharmacological therapy for dystrophinopathies comprises glucocorticoids (prednisone, prednisolone, and deflazacort), vamorolone, and ataluren. However, angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs), and β-blockers are the first-line to prevent dilated cardiomyopathy in dystrophinopathy patients. Duchenne muscular dystrophy gene therapy strategies involve gene transfer, exon skipping, exon reframing, and CRISPR gene editing. Eteplirsen, an antisense-oligonucleotide drug for skipping exon 51 from the Dystrophin gene, is available on the market, which may help up to 14% of Duchenne muscular dystrophy patients. There are various FDA-approved exon skipping drugs including ExonDys-51 for exon 51, VyonDys-53 and Viltolarsen for exon 53 and AmonDys-45 for exon 45 skipping. Other antisense oligonucleotide drugs in the pipeline include casimersen for exon 45, suvodirsen for exon 51, and golodirsen for exon 53 skipping. Advances in the diagnosis and therapy of dystrophinopathies offer new perspectives for their early discovery and care.
Collapse
Affiliation(s)
- Fawzy A. Saad
- Department of Gene Therapy, Saad Pharmaceuticals, Juhkentali 8, 10132 Tallinn, Estonia
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, Pisa University School of Medicine, Via Paradisa 2, 56100 Pisa, Italy;
| | - Corrado Angelini
- Department of Neurosciences, Padova University School of Medicine, Via Giustiniani 5, 35128 Padova, Italy;
| |
Collapse
|
9
|
Chey YCJ, Arudkumar J, Aartsma-Rus A, Adikusuma F, Thomas PQ. CRISPR applications for Duchenne muscular dystrophy: From animal models to potential therapies. WIREs Mech Dis 2023; 15:e1580. [PMID: 35909075 PMCID: PMC10078488 DOI: 10.1002/wsbm.1580] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
CRISPR gene-editing technology creates precise and permanent modifications to DNA. It has significantly advanced our ability to generate animal disease models for use in biomedical research and also has potential to revolutionize the treatment of genetic disorders. Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disease that could potentially benefit from the development of CRISPR therapy. It is commonly associated with mutations that disrupt the reading frame of the DMD gene that encodes dystrophin, an essential scaffolding protein that stabilizes striated muscles and protects them from contractile-induced damage. CRISPR enables the rapid generation of various animal models harboring mutations that closely simulates the wide variety of mutations observed in DMD patients. These models provide a platform for the testing of sequence-specific interventions like CRISPR therapy that aim to reframe or skip DMD mutations to restore functional dystrophin expression. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,CSIRO Synthetic Biology Future Science Platform, Canberra, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
10
|
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: 1.5] [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
|
11
|
CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives. Cells 2022; 11:cells11192964. [PMID: 36230926 PMCID: PMC9564082 DOI: 10.3390/cells11192964] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease arising from loss-of-function mutations in the dystrophin gene and characterized by progressive muscle degeneration, respiratory insufficiency, cardiac failure, and premature death by the age of thirty. Albeit DMD is one of the most common types of fatal genetic diseases, there is no curative treatment for this devastating disorder. In recent years, gene editing via the clustered regularly interspaced short palindromic repeats (CRISPR) system has paved a new path toward correcting pathological mutations at the genetic source, thus enabling the permanent restoration of dystrophin expression and function throughout the musculature. To date, the therapeutic benefits of CRISPR genome-editing systems have been successfully demonstrated in human cells, rodents, canines, and piglets with diverse DMD mutations. Nevertheless, there remain some nonignorable challenges to be solved before the clinical application of CRISPR-based gene therapy. Herein, we provide an overview of therapeutic CRISPR genome-editing systems, summarize recent advancements in their applications in DMD contexts, and discuss several potential obstacles lying ahead of clinical translation.
Collapse
|
12
|
Wang J, Fan Q, Yu T, Zhang Y. Identifying the hub genes for Duchenne muscular dystrophy and Becker muscular dystrophy by weighted correlation network analysis. BMC Genom Data 2021; 22:57. [PMID: 34922439 PMCID: PMC8684282 DOI: 10.1186/s12863-021-01014-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022] Open
Abstract
Background The goal of this study is to identify the hub genes for Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) via weighted correlation network analysis (WGCNA). Methods The gene expression profile of vastus lateralis biopsy samples obtained in 17 patients with DMD, 11 patients with BMD and 6 healthy individuals was downloaded from the Gene Expression Omnibus (GEO) database (GSE109178). After obtaining different expressed genes (DEGs) via GEO2R, WGCNA was conducted using R package, modules and genes that highly associated with DMD, BMD, and their age or pathology were screened. Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) enrichment analysis and protein–protein interaction (PPI) network analysis were also conducted. Hub genes and highly correlated clustered genes were identified using Search Tool for the Retrieval of Interacting Genes (STRING) and Cystoscape software. Results One thousand four hundred seventy DEGs were identified between DMD and control, with 1281 upregulated and 189 downregulated DEGs. Four hundred and twenty DEGs were found between BMD and control, with 157 upregulated and 263 upregulated DEGs. Fourteen modules with different colors were identified for DMD vs control, and 7 modules with different colors were identified for BMD vs control. Ten hub genes were summarized for DMD and BMD respectively, 5 hub genes were summarized for BMD age, 5 and 3 highly correlated clustered genes were summarized for DMD age and BMD pathology, respectively. In addition, 20 GO enrichments were found to be involved in DMD, 3 GO enrichments were found to be involved in BMD, 3 GO enrichments were found to be involved in BMD age. Conclusion In DMD, several hub genes were identified: C3AR1, TLR7, IRF8, FYB and CD33(immune and inflammation associated genes), TYROBP, PLEK, AIF1(actin reorganization associated genes), LAPTM5 and NT5E(cell death and arterial calcification associated genes, respectively). In BMD, a number of hub genes were identified: LOX, ELN, PLEK, IKZF1, CTSK, THBS2, ADAMTS2, COL5A1(extracellular matrix associated genes), BCL2L1 and CDK2(cell cycle associated genes).
Collapse
|
13
|
Becker muscular dystrophy: case report, review of the literature, and analysis of differentially expressed hub genes. Neurol Sci 2021; 43:243-253. [PMID: 34731335 DOI: 10.1007/s10072-021-05499-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/21/2021] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Becker muscular dystrophy (BMD) is a genetic and progressive neuromuscular disease caused by mutations in the dystrophin gene with no available cure. A case report and comprehensive review of BMD cases aim to provide important clues for early diagnosis and implications for clinical practice. Genes and pathways identified from microarray data of muscle samples from patients with BMD help uncover the potential mechanism and provide novel therapeutic targets for dystrophin-deficient muscular dystrophies. METHODS We describe a BMD family with a 10-year-old boy as the proband and reviewed BMD cases from PubMed. Datasets from the Gene Expression Omnibus database were downloaded and integrated with the online software. RESULTS The systematic review revealed the clinical manifestations and mutation points of the dystrophin gene. Gene ontology analysis showed that extracellular matrix organization and extracellular structure organization with enrichment of upregulated genes coexist in three datasets. We present the first report of TUBA1A involvement in the development of BMD/Duchenne muscular dystrophy (DMD). DISCUSSION This study provides important implications for clinical practice, uncovering the potential mechanism of the progress of BMD/DMD, and provided new therapeutic targets.
Collapse
|
14
|
Sheikh O, Yokota T. Developing DMD therapeutics: a review of the effectiveness of small molecules, stop-codon readthrough, dystrophin gene replacement, and exon-skipping therapies. Expert Opin Investig Drugs 2021; 30:167-176. [PMID: 33393390 DOI: 10.1080/13543784.2021.1868434] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder caused by mutations in the dystrophin (DMD) gene. Most patients die from respiratory failure or cardiomyopathy. There are significant unmet needs for treatments for DMD as the standard of care is principally limited to symptom relief through treatments including steroids. AREAS COVERED This review summarizes safety and efficacy in promising areas of DMD therapeutics - small molecules, stop codon readthrough, gene replacement, and exon skipping - under clinical examination from 2015-2020 as demonstrated in the NIH Clinical Trials and PubMed search engines. EXPERT OPINION Currently, steroids persist as the most accessible medicine for DMD. Stop-codon readthrough, gene replacement, and exon-skipping therapies all aim to restore dystrophin expression. Of these strategies, gene replacement therapy has recently gained momentum while exon-skipping retains great traction. The FDA approval of three exon-skipping antisense oligonucleotides illustrate this regulatory momentum, though the effectiveness and sequence design of eteplirsen remain controversial. Cell-penetrating peptides promise to more efficaciously treat DMD-related cardiomyopathy.The recent success of antisense therapies, however, poses major regulatory challenges. To fully realize the benefits of exon-skipping, including cocktail oligonucleotide-mediated multiple exon-skipping and oligonucleotide drugs for very rare mutations, regulatory challenges need to be addressed in coordination with scientific advances.
Collapse
Affiliation(s)
- Omar Sheikh
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta , Edmonton, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta , Edmonton, Canada
| |
Collapse
|
15
|
Gibbs EM, Barthélémy F, Douine ED, Hardiman NC, Shieh PB, Khanlou N, Crosbie RH, Nelson SF, Miceli MC. Large in-frame 5' deletions in DMD associated with mild Duchenne muscular dystrophy: Two case reports and a review of the literature. Neuromuscul Disord 2019; 29:863-873. [PMID: 31672265 PMCID: PMC7092699 DOI: 10.1016/j.nmd.2019.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 10/26/2022]
Abstract
Duchenne muscular dystrophy is caused by mutations in the dystrophin-encoding DMD gene. While Duchenne is most commonly caused by large intragenic deletions that cause frameshift and complete loss of dystrophin expression, in-frame deletions in DMD can result in the expression of internally truncated dystrophin proteins and may be associated with a milder phenotype. In this study, we describe two individuals with large in-frame 5' deletions (exon 3-23 and exon 3-28) that remove the majority of the N-terminal region, including part of the actin binding and central rod domains. Both patients had progressive muscle weakness during childhood but are observed to have a relatively mild disease course compared to typical Duchenne. We show that in muscle biopsies from both patients, truncated dystrophin is expressed at the sarcolemma. We have additionally developed a patient-specific fibroblast-derived cell model, which can be inducibly reprogrammed to form myotubes that largely recapitulate biopsy findings for the patient with the exon 3-23 deletion, providing a culture model for future investigation of this unusual case. We discuss these mutations in the context of previously reported 5' in-frame DMD deletions and relevant animal models, and review the spectrum of phenotypes associated with these deletions.
Collapse
Affiliation(s)
- Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA
| | - Florian Barthélémy
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Emilie D Douine
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Natalie C Hardiman
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Perry B Shieh
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, 90095, USA
| | - Negar Khanlou
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, 90095, USA
| | - M Carrie Miceli
- Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
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: 5.8] [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
|
17
|
Wells DJ. What is the level of dystrophin expression required for effective therapy of Duchenne muscular dystrophy? J Muscle Res Cell Motil 2019; 40:141-150. [PMID: 31289969 DOI: 10.1007/s10974-019-09535-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked muscle wasting disease. The disease is due to mutations in the DMD gene that encodes for a large intracellular protein called dystrophin. Dystrophin plays a critical role in linking the internal cytoskeleton of the striated muscle cell with the extracellular matrix as well as having cell signalling functions. In its absence muscle contraction is associated with cycles of damage, repair, inflammation and fibrosis with eventual loss of muscle and replacement with fat. Experiments in animal models of DMD have generated a number of different approaches to the induction of dystrophin including viral vector mediated delivery of a recombinant dystrophin gene, antisense oligonucleotide mediated exon-skipping to restore the open reading frame in the dystrophin mRNA, read-through of premature stop mutations, genome modification using CRISPR-Cas9 or cell based transfer of a functional dystrophin gene. In all cases, it will be important to understand how much dystrophin expression is required for a clinically effective therapy and this review examines the data from humans and animal models to estimate the percentage of endogenous dystrophin that is likely to have significant clinical benefit. While there are a number of important caveats to consider, including the appropriate outcome measures, this review suggests that approximately 20% of endogenous levels uniformly distributed within the skeletal muscles and the heart may be sufficient to largely prevent disease progression.
Collapse
Affiliation(s)
- Dominic J Wells
- Neuromuscular Diseases Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
18
|
Nakamura A. Mutation-Based Therapeutic Strategies for Duchenne Muscular Dystrophy: From Genetic Diagnosis to Therapy. J Pers Med 2019; 9:jpm9010016. [PMID: 30836656 PMCID: PMC6462977 DOI: 10.3390/jpm9010016] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 02/06/2023] Open
Abstract
Duchenne and Becker muscular dystrophy (DMD/BMD) are X-linked muscle disorders caused by mutations of the DMD gene, which encodes the subsarcolemmal protein dystrophin. In DMD, dystrophin is not expressed due to a disruption in the reading frame of the DMD gene, resulting in a severe phenotype. Becker muscular dystrophy exhibits a milder phenotype, having mutations that maintain the reading frame and allow for the production of truncated dystrophin. To date, various therapeutic approaches for DMD have been extensively developed. However, the pathomechanism is quite complex despite it being a single gene disorder, and dystrophin is expressed not only in a large amount of skeletal muscle but also in cardiac, vascular, intestinal smooth muscle, and nervous system tissue. Thus, the most appropriate therapy would be complementation or restoration of dystrophin expression, such as gene therapy using viral vectors, readthrough therapy, or exon skipping therapy. Among them, exon skipping therapy with antisense oligonucleotides can restore the reading frame and yield the conversion of a severe phenotype to one that is mild. In this paper, I present the significance of molecular diagnosis and the development of mutation-based therapeutic strategies to complement or restore dystrophin expression.
Collapse
Affiliation(s)
- Akinori Nakamura
- Department of Neurology, National Hospital Organization, Matsumoto Medical Center, 2-20-30 Murai-machi Minami, Matsumoto 399-8701, Japan.
- Third Department of Medicine, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan.
| |
Collapse
|
19
|
Multiple Exon Skipping in the Duchenne Muscular Dystrophy Hot Spots: Prospects and Challenges. J Pers Med 2018; 8:jpm8040041. [PMID: 30544634 PMCID: PMC6313462 DOI: 10.3390/jpm8040041] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/24/2018] [Accepted: 12/04/2018] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), a fatal X-linked recessive disorder, is caused mostly by frame-disrupting, out-of-frame deletions in the dystrophin (DMD) gene. Antisense oligonucleotide-mediated exon skipping is a promising therapy for DMD. Exon skipping aims to convert out-of-frame mRNA to in-frame mRNA and induce the production of internally-deleted dystrophin as seen in the less severe Becker muscular dystrophy. Currently, multiple exon skipping has gained special interest as a new therapeutic modality for this approach. Previous retrospective database studies represented a potential therapeutic application of multiple exon skipping. Since then, public DMD databases have become more useful with an increase in patient registration and advances in molecular diagnosis. Here, we provide an update on DMD genotype-phenotype associations using a global DMD database and further provide the rationale for multiple exon skipping development, particularly for exons 45–55 skipping and an emerging therapeutic concept, exons 3–9 skipping. Importantly, this review highlights the potential of multiple exon skipping for enabling the production of functionally-corrected dystrophin and for treating symptomatic patients not only with out-of-frame deletions but also those with in-frame deletions. We will also discuss prospects and challenges in multiple exon skipping therapy, referring to recent progress in antisense chemistry and design, as well as disease models.
Collapse
|
20
|
Personalized gene and cell therapy for Duchenne Muscular Dystrophy. Neuromuscul Disord 2018; 28:803-824. [DOI: 10.1016/j.nmd.2018.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 01/09/2023]
|
21
|
Genetic and pharmacological regulation of the endocannabinoid CB1 receptor in Duchenne muscular dystrophy. Nat Commun 2018; 9:3950. [PMID: 30262909 PMCID: PMC6160489 DOI: 10.1038/s41467-018-06267-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 08/22/2018] [Indexed: 12/14/2022] Open
Abstract
The endocannabinoid system refers to a widespread signaling system and its alteration is implicated in a growing number of human diseases. However, the potential role of endocannabinoids in skeletal muscle disorders remains unknown. Here we report the role of the endocannabinoid CB1 receptors in Duchenne's muscular dystrophy. In murine and human models, CB1 transcripts show the highest degree of expression at disease onset, and then decline overtime. Similar changes are observed for PAX7, a key regulator of muscle stem cells. Bioinformatics and biochemical analysis reveal that PAX7 binds and upregulates the CB1 gene in dystrophic more than in healthy muscles. Rimonabant, an antagonist of CB1, promotes human satellite cell differentiation in vitro, increases the number of regenerated myofibers, and prevents locomotor impairment in dystrophic mice. In conclusion, our study uncovers a PAX7-CB1 cross talk potentially exacerbating DMD and highlights the role of CB1 receptors as target for potential therapies.
Collapse
|
22
|
Yuan R, Yi J, Xie Z, Zheng Y, Han M, Hou Y, Wang Z, Yuan Y. Genotype-phenotype correlation in Becker muscular dystrophy in Chinese patients. J Hum Genet 2018; 63:1041-1048. [PMID: 29976999 DOI: 10.1038/s10038-018-0480-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 05/23/2018] [Accepted: 06/01/2018] [Indexed: 12/26/2022]
Abstract
Large deletions and duplications are the most frequent causative mutations in Becker muscular dystrophy (BMD), but genetic profile varied greatly among reports. We performed a comprehensive molecular investigation in 95 Chinese BMD patients. All patients were divided into three subtypes: normal muscle strength (type 1) in 18 cases, quadriceps myopathy (type 2) in 20 cases, and limb-girdle weakness (type 3) in 57 cases. Nineteen cases (20.0%) had small mutations and 76 cases (80.0%) had major rearrangements, including 67 cases (70.5%) of exonic deletions and 9 cases (9.5%) of exonic duplications. We identified 50 cases (65.8%) of in-frame mutations, and 26 cases (34.2%) of frame-shift mutations. The frequency of deletion in exons 13-19 was 30.6% in type 1 patients, 9.7% in type 2 patients, and 10.4% in type 3 patients. The frequency of deletion in exons 45-55 was 28.6% in type 1 patients, 40.8% in type 2, and 50.0% in type 3 patients. All major rearrangements of DMD gene in type 1 patients were also observed in type 3 patients. Our study suggested that frame-shift mutation was not rare in Chinese BMD patients. Although no difference was observed on the forms of DMD gene mutations among the three types of patients, the mutation in proximal region of DMD gene has higher frequency for patients without weakness. Effect of exon skipping for DMD depends on the size and location of the mutation. Additional studies are required to determine whether exon-skipping strategies in proximal region of DMD gene could yield more functional dystrophin.
Collapse
Affiliation(s)
- Ruiyi Yuan
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China.,Haverford Collage, Haverford, PA, 19041, USA
| | - Junfei Yi
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China
| | - Zhiying Xie
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China
| | - Yimeng Zheng
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China
| | - Miao Han
- Department of Neurology, First Hospital, Henan University, Kaifeng Shi, China
| | - Yue Hou
- Department of Neurology, Traditional Medicine Hospital, Guangzhou University, Guangzhou, China
| | - Zhaoxia Wang
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China
| | - Yun Yuan
- Department of Neurology, First Hospital, Peking University, 100034, Beijing, China.
| |
Collapse
|
23
|
Rodrigues M, Echigoya Y, Fukada SI, Yokota T. Current Translational Research and Murine Models For Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 3:29-48. [PMID: 27854202 PMCID: PMC5271422 DOI: 10.3233/jnd-150113] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder characterized by progressive muscle degeneration. Mutations in the DMD gene result in the absence of dystrophin, a protein required for muscle strength and stability. Currently, there is no cure for DMD. Since murine models are relatively easy to genetically manipulate, cost effective, and easily reproducible due to their short generation time, they have helped to elucidate the pathobiology of dystrophin deficiency and to assess therapies for treating DMD. Recently, several murine models have been developed by our group and others to be more representative of the human DMD mutation types and phenotypes. For instance, mdx mice on a DBA/2 genetic background, developed by Fukada et al., have lower regenerative capacity and exhibit very severe phenotype. Cmah-deficient mdx mice display an accelerated disease onset and severe cardiac phenotype due to differences in glycosylation between humans and mice. Other novel murine models include mdx52, which harbors a deletion mutation in exon 52, a hot spot region in humans, and dystrophin/utrophin double-deficient (dko), which displays a severe dystrophic phenotype due the absence of utrophin, a dystrophin homolog. This paper reviews the pathological manifestations and recent therapeutic developments in murine models of DMD such as standard mdx (C57BL/10), mdx on C57BL/6 background (C57BL/6-mdx), mdx52, dystrophin/utrophin double-deficient (dko), mdxβgeo, Dmd-null, humanized DMD (hDMD), mdx on DBA/2 background (DBA/2-mdx), Cmah-mdx, and mdx/mTRKO murine models.
Collapse
Affiliation(s)
- Merryl Rodrigues
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Yusuke Echigoya
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - So-Ichiro Fukada
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada.,Muscular Dystrophy Canada Research Chair, Edmonton, Alberta, Canada
| |
Collapse
|
24
|
Clinical Utility Gene Card for: Becker muscular dystrophy. Eur J Hum Genet 2018; 26:1065-1071. [PMID: 29467387 DOI: 10.1038/s41431-017-0064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 09/10/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022] Open
|
25
|
Aslesh T, Maruyama R, Yokota T. Skipping Multiple Exons to Treat DMD-Promises and Challenges. Biomedicines 2018; 6:E1. [PMID: 29301272 PMCID: PMC5874658 DOI: 10.3390/biomedicines6010001] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disorder caused by mutations in the DMD gene. Antisense-mediated exon-skipping is a promising therapeutic strategy that makes use of synthetic nucleic acids to skip frame-disrupting exon(s) and allows for short but functional protein expression by restoring the reading frame. In 2016, the U.S. Food and Drug Administration (FDA) approved eteplirsen, which skips DMD exon 51 and is applicable to approximately 13% of DMD patients. Multiple exon skipping, which is theoretically applicable to 80-90% of DMD patients in total, have been demonstrated in animal models, including dystrophic mice and dogs, using cocktail antisense oligonucleotides (AOs). Although promising, current drug approval systems pose challenges for the use of a cocktail AO. For example, both exons 6 and 8 need to be skipped to restore the reading frame in dystrophic dogs. Therefore, the cocktail of AOs targeting these exons has a combined therapeutic effect and each AO does not have a therapeutic effect by itself. The current drug approval system is not designed to evaluate such circumstances, which are completely different from cocktail drug approaches in other fields. Significant changes are needed in the drug approval process to promote the cocktail AO approach.
Collapse
Affiliation(s)
- Tejal Aslesh
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 8812-112 St. Edmonton, AB T6G 2H7, Canada.
| | - Rika Maruyama
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 8812-112 St. Edmonton, AB T6G 2H7, Canada.
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 8812-112 St. Edmonton, AB T6G 2H7, Canada.
- The Friends of Garrett Cumming Research and Muscular Dystrophy Canada HM Toupin Neurological Science Research Chair, 8812-112 St. Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
26
|
A rare subclinical or mild type of Becker muscular dystrophy caused by a single exon 48 deletion of the dystrophin gene. J Appl Genet 2017; 58:343-347. [PMID: 28247318 PMCID: PMC5509810 DOI: 10.1007/s13353-017-0391-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 01/10/2017] [Accepted: 01/30/2017] [Indexed: 01/20/2023]
Abstract
In the material of 227 families with Becker muscular dystrophy (BMD), we found nine non-consanguineous families with 17 male individuals carrying a rare mutation—a single exon 48 deletion of the dystrophin gene—who were affected with a very mild or subclinical form of BMD. They were usually detected thanks to accidental findings of elevated serum creatine phosphokinase (sCPK). A thorough clinical analysis of the carriers, both children (12) and adults (5), revealed in some of them muscle hypotonia (10/17) and/or very mild muscle weakness (9/17), as well as decreased tendon reflexes (6/17). Adults, apart from very mild muscle weakness and calf hypertrophy in some, had no significant abnormalities on neurological assessments and had good exercise tolerance. Parents of the children carriers of the exon 48 deletion are usually unaware of their children being affected, and possibly at risk of developing life-threatening cardiomyopathy. The same concerns the adult male carriers. Therefore, the authors postulate undertaking preventive measures such as cascade screening of the relatives of the probands. Newborn screening programmes of Duchenne muscular dystrophy (DMD)/BMD based on sCPK marked increase may be considered.
Collapse
|
27
|
|
28
|
Bello L, Campadello P, Barp A, Fanin M, Semplicini C, Sorarù G, Caumo L, Calore C, Angelini C, Pegoraro E. Functional changes in Becker muscular dystrophy: implications for clinical trials in dystrophinopathies. Sci Rep 2016; 6:32439. [PMID: 27582364 PMCID: PMC5007528 DOI: 10.1038/srep32439] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/05/2016] [Indexed: 01/16/2023] Open
Abstract
We performed a 1-year longitudinal study of Six Minute Walk Test (6MWT), North Star Ambulatory Assessment (NSAA), and timed function tests in Becker muscular dystrophy (BMD). Skeletal muscle dystrophin was quantified by immunoblot. We grouped deletions ending on exon 45 ("del 45-x", n = 28) or 51 ("del x-51", n = 10); isolated exon 48 deletion ("del 48", n = 10); and other mutations (n = 21). Only patients in the "del 45-x" or "other" groups became non-ambulatory (n = 5, log-rank p = n.s.) or unable to run (n = 22, p < 0.001). All measures correlated positively with dystrophin quantity and negatively with age, and were significantly more impaired in the "del 45-x" and "other" groups. After one year, NSAA score decreased significantly (-0.9 ± 1.6, p < 0.001); in the "del 45-x" group, both NSAA (-1.3 ± 1.7, p = 0.001) and 6MWT (-12 ± 31 m, p = 0.059) decreased. We conclude that patients with "del x-51" or "del 48" mutations have mild or asymptomatic BMD, while "del 45-x" mutations cause comparatively severe weakness, and functional deterioration in 1 year. Furthermore, exon 51 skipping could be more effective than exon 45 skipping in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Paola Campadello
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Andrea Barp
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Marina Fanin
- Department of Neurosciences, University of Padova, Padova, Italy
| | | | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Luca Caumo
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Chiara Calore
- Department of Cardiac, Thoracic and Vascular Sciences, Cardiology Clinic, University of Padova, Padova, Italy
| | - Corrado Angelini
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
29
|
Miskew Nichols B, Aoki Y, Kuraoka M, Lee JJA, Takeda S, Yokota T. Multi-exon Skipping Using Cocktail Antisense Oligonucleotides in the Canine X-linked Muscular Dystrophy. J Vis Exp 2016. [PMID: 27285612 PMCID: PMC4927712 DOI: 10.3791/53776] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is one of the most common lethal genetic diseases worldwide, caused by mutations in the dystrophin (DMD) gene. Exon skipping employs short DNA/RNA-like molecules called antisense oligonucleotides (AONs) that restore the reading frame and produce shorter but functional proteins. However, exon skipping therapy faces two major hurdles: limited applicability (up to only 13% of patients can be treated with a single AON drug), and uncertain function of truncated proteins. These issues were addressed with a cocktail AON approach. While approximately 70% of DMD patients can be treated by single exon skipping (all exons combined), one could potentially treat more than 90% of DMD patients if multiple exon skipping using cocktail antisense drugs can be realized. The canine X-linked muscular dystrophy (CXMD) dog model, whose phenotype is more similar to human DMD patients, was used to test the systemic efficacy and safety of multi-exon skipping of exons 6 and 8. The CXMD dog model harbors a splice site mutation in intron 6, leading to a lack of exon 7 in dystrophin mRNA. To restore the reading frame in CXMD requires multi-exon skipping of exons 6 and 8; therefore, CXMD is a good middle-sized animal model for testing the efficacy and safety of multi-exon skipping. In the current study, a cocktail of antisense morpholinos targeting exon 6 and exon 8 was designed and it restored dystrophin expression in body-wide skeletal muscles. Methods for transfection/injection of cocktail oligos and evaluation of the efficacy and safety of multi-exon skipping in the CXMD dog model are presented.
Collapse
Affiliation(s)
- Bailey Miskew Nichols
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry
| | - Joshua J A Lee
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry;
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta Faculty of Medicine and Dentistry;
| |
Collapse
|
30
|
Abstract
The dystrophin complex stabilizes the plasma membrane of striated muscle cells. Loss of function mutations in the genes encoding dystrophin, or the associated proteins, trigger instability of the plasma membrane, and myofiber loss. Mutations in dystrophin have been extensively cataloged, providing remarkable structure-function correlation between predicted protein structure and clinical outcomes. These data have highlighted dystrophin regions necessary for in vivo function and fueled the design of viral vectors and now, exon skipping approaches for use in dystrophin restoration therapies. However, dystrophin restoration is likely more complex, owing to the role of the dystrophin complex as a broad cytoskeletal integrator. This review will focus on dystrophin restoration, with emphasis on the regions of dystrophin essential for interacting with its associated proteins and discuss the structural implications of these approaches.
Collapse
Affiliation(s)
- Quan Q Gao
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, Illinois, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, Chicago, Illinois, USA
| |
Collapse
|
31
|
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy are caused by mutations in the dystrophin-encoding DMD gene. Large deletions and duplications are most common, but small mutations have been found as well. Having a correct diagnosis is important for family planning and providing proper care to patients according to published guidelines. With mutation-specific therapies under development for DMD, a correct diagnosis is now also important for assessing whether patients are eligible for treatments. This review discusses different mutations causing DMD, diagnostic techniques available for making a genetic diagnosis for children suspected of DMD and the importance of having a specific genetic diagnosis in the context of emerging genetic therapies for DMD.
Collapse
Affiliation(s)
- Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands,John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Ieke B Ginjaar
- Laboratory for Diagnostics Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| |
Collapse
|
32
|
Aartsma-Rus A, Ginjaar IB, Bushby K. The importance of genetic diagnosis for Duchenne muscular dystrophy. J Med Genet 2016; 53:145-51. [PMID: 26754139 PMCID: PMC4789806 DOI: 10.1136/jmedgenet-2015-103387] [Citation(s) in RCA: 215] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/10/2015] [Indexed: 12/18/2022]
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy are caused by mutations in the dystrophin-encoding DMD gene. Large deletions and duplications are most common, but small mutations have been found as well. Having a correct diagnosis is important for family planning and providing proper care to patients according to published guidelines. With mutation-specific therapies under development for DMD, a correct diagnosis is now also important for assessing whether patients are eligible for treatments. This review discusses different mutations causing DMD, diagnostic techniques available for making a genetic diagnosis for children suspected of DMD and the importance of having a specific genetic diagnosis in the context of emerging genetic therapies for DMD.
Collapse
Affiliation(s)
- Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Ieke B Ginjaar
- Laboratory for Diagnostics Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre and MRC Centre for Neuromuscular diseases, Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| |
Collapse
|
33
|
McGreevy JW, Hakim CH, McIntosh MA, Duan D. Animal models of Duchenne muscular dystrophy: from basic mechanisms to gene therapy. Dis Model Mech 2015; 8:195-213. [PMID: 25740330 PMCID: PMC4348559 DOI: 10.1242/dmm.018424] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disorder. It is caused by loss-of-function mutations in the dystrophin gene. Currently, there is no cure. A highly promising therapeutic strategy is to replace or repair the defective dystrophin gene by gene therapy. Numerous animal models of DMD have been developed over the last 30 years, ranging from invertebrate to large mammalian models. mdx mice are the most commonly employed models in DMD research and have been used to lay the groundwork for DMD gene therapy. After ~30 years of development, the field has reached the stage at which the results in mdx mice can be validated and scaled-up in symptomatic large animals. The canine DMD (cDMD) model will be excellent for these studies. In this article, we review the animal models for DMD, the pros and cons of each model system, and the history and progress of preclinical DMD gene therapy research in the animal models. We also discuss the current and emerging challenges in this field and ways to address these challenges using animal models, in particular cDMD dogs.
Collapse
Affiliation(s)
- Joe W McGreevy
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Mark A McIntosh
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
34
|
|
35
|
Development of multiexon skipping antisense oligonucleotide therapy for Duchenne muscular dystrophy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:402369. [PMID: 23984357 PMCID: PMC3747431 DOI: 10.1155/2013/402369] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/18/2013] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an incurable, X-linked progressive muscle degenerative disorder that results from the absence of dystrophin protein and leads to premature death in affected individuals due to respiratory and/or cardiac failure typically by age of 30. Very recently the exciting prospect of an effective oligonucleotide therapy has emerged which restores dystrophin protein expression to affected tissues in DMD patients with highly promising data from a series of clinical trials. This therapeutic approach is highly mutation specific and thus is personalised. Therefore DMD has emerged as a model genetic disorder for understanding and overcoming of the challenges of developing personalised genetic medicines. One of the greatest weaknesses of the current oligonucleotide approach is that it is a mutation-specific therapy. To address this limitation, we have recently demonstrated that exons 45–55 skipping therapy has the potential to treat clusters of mutations that cause DMD, which could significantly reduce the number of compounds that would need to be developed in order to successfully treat all DMD patients. Here we discuss and review the latest preclinical work in this area as well as a variety of accompanying issues, including efficacy and potential toxicity of antisense oligonucleotides, prior to human clinical trials.
Collapse
|
36
|
Silvestri NJ, Wolfe GI. Asymptomatic/pauci-symptomatic creatine kinase elevations (hyperckemia). Muscle Nerve 2013; 47:805-15. [DOI: 10.1002/mus.23755] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2012] [Indexed: 11/06/2022]
Affiliation(s)
- Nicholas J. Silvestri
- Department of Neurology; University at Buffalo, Buffalo General Medical Center; 100 High Street Buffalo New York 14203-1126 USA
| | - Gil I. Wolfe
- Department of Neurology; University at Buffalo, Buffalo General Medical Center; 100 High Street Buffalo New York 14203-1126 USA
| |
Collapse
|
37
|
Li F, Li Y, Cui K, Li C, Chen W, Gao J, Zhu Y, Zeng C, Li S. Detection of pathogenic mutations and the mechanism of a rare chromosomal rearrangement in a Chinese family with Becker muscular dystrophy. Clin Chim Acta 2012; 414:20-5. [PMID: 22910583 DOI: 10.1016/j.cca.2012.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 07/21/2012] [Accepted: 08/06/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE The objectives of this research are to genetically diagnose a family with Becker muscular dystrophy (BMD), to explore the molecular mechanism of the disease, and to predict the possibility of BMD development in two individuals who have not yet reached the age of onset (young individuals). METHODS The multiplex polymerase chain reaction was first employed to screen dystrophin (DMD) gene deletions, and the locations of deletion breakpoints were identified using the Sequenom platform and long-range PCR. Sanger sequencing was then performed for the undeleted exons. RESULTS All BMD patients and a young individual carry a deletion spanning exons 45 to 53 and an unreported missense mutation on exon 11 of the DMD gene. This point mutation was screened in 412 healthy individuals and heterozygous genotype was found in two females. Determination of deletion breakpoints demonstrated a 330-kb deletion and there was a 9-bp insertion between the breakpoints. This 9-bp could match a reference sequence located within the deleted region. CONCLUSIONS Two mutations of the DMD gene coexist in this family. One young child has a high disease risk. Pathogenic potential of the point mutation requires further investigation. The rare chromosomal rearrangement may be caused by short-nucleotide sequence capture or other unknown mechanisms.
Collapse
Affiliation(s)
- Feifei Li
- Laboratory of Disease Genomics and Individualized Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Bodywide skipping of exons 45-55 in dystrophic mdx52 mice by systemic antisense delivery. Proc Natl Acad Sci U S A 2012; 109:13763-8. [PMID: 22869723 DOI: 10.1073/pnas.1204638109] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), the commonest form of muscular dystrophy, is caused by lack of dystrophin. One of the most promising therapeutic approaches is antisense-mediated elimination of frame-disrupting mutations by exon skipping. However, this approach faces two major hurdles: limited applicability of each individual target exon and uncertain function and stability of each resulting truncated dystrophin. Skipping of exons 45-55 at the mutation hotspot of the DMD gene would address both issues. Theoretically it could rescue more than 60% of patients with deletion mutations. Moreover, spontaneous deletions of this specific region are associated with asymptomatic or exceptionally mild phenotypes. However, such multiple exon skipping of exons 45-55 has proved technically challenging. We have therefore designed antisense oligo (AO) morpholino mixtures to minimize self- or heteroduplex formation. These were tested as conjugates with cell-penetrating moieties (vivo-morpholinos). We have tested the feasibility of skipping exons 45-55 in H2K-mdx52 myotubes and in mdx52 mice, which lack exon 52. Encouragingly, with mixtures of 10 AOs, we demonstrated skipping of all 10 exons in vitro, in H2K-mdx52 myotubes and on intramuscular injection into mdx52 mice. Moreover, in mdx52 mice in vivo, systemic injections of 10 AOs induced extensive dystrophin expression at the subsarcolemma in skeletal muscles throughout the body, producing up to 15% of wild-type dystrophin protein levels, accompanied by improved muscle strength and histopathology without any detectable toxicity. This is a unique successful demonstration of effective rescue by exon 45-55 skipping in a dystrophin-deficient animal model.
Collapse
|
39
|
Assessing pathogenicity for novel mutation/sequence variants: the value of healthy older individuals. Neuromolecular Med 2012; 14:281-4. [PMID: 22707356 PMCID: PMC3505535 DOI: 10.1007/s12017-012-8186-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 06/01/2012] [Indexed: 11/06/2022]
Abstract
Improvement in DNA technology is increasingly revealing unexpected/unknown mutations in healthy persons and generating anxiety due to their still unknown health consequences. We report a 44-year-old healthy father of a 10-year-old daughter with bilateral coloboma and hearing loss, but without muscle weakness, in whom a whole-genome CGH revealed a deletion of exons 38–44 in the dystrophin gene. This mutation was inherited from her asymptomatic father, who was further clinically and molecularly evaluated for prognosis and genetic counseling (GC). This deletion was never identified by us in 982 Duchenne/Becker patients. To assess whether the present case represents a rare case of non-penetrance, and aiming to obtain more information for prognosis and GC, we suggested that healthy older relatives submit their DNA for analysis, to which several complied. Mutation analysis revealed that his mother, brother, and 56-year-old maternal uncle also carry the 38–44 deletion, suggesting it an unlikely cause of muscle weakness. Genome sequencing will disclose mutations and variants whose health impact are still unknown, raising important problems in interpreting results, defining prognosis, and discussing GC. We suggest that, in addition to family history, keeping the DNA of older relatives could be very informative, in particular for those interested in having their genome sequenced.
Collapse
|
40
|
Goyenvalle A, Wright J, Babbs A, Wilkins V, Garcia L, Davies KE. Engineering multiple U7snRNA constructs to induce single and multiexon-skipping for Duchenne muscular dystrophy. Mol Ther 2012; 20:1212-21. [PMID: 22354379 DOI: 10.1038/mt.2012.26] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disorder caused by mutations in the dystrophin gene. Antisense-mediated exon skipping is one of the most promising approaches for the treatment of DMD but still faces personalized medicine challenges as different mutations found in DMD patients require skipping of different exons. However, 70% of DMD patients harbor dystrophin gene deletions in a mutation-rich area or "hot-spot" in the central genomic region. In this study, we have developed 11 different U7 small-nuclear RNA, to shuttle antisense sequences designed to mask key elements involved in the splicing of exons 45 to 55. We demonstrate that these constructs induce efficient exon skipping both in vitro in DMD patients' myoblasts and in vivo in human DMD (hDMD) mice and that they can be combined into a single vector to achieve a multi skipping of at least 3 exons. These very encouraging results provide proof of principle that efficient multiexon-skipping can be achieved using adeno-associated viral (AAV) vectors encoding multiple U7 small-nuclear RNAs (U7snRNAs), offering therefore very promising tools for clinical treatment of DMD.
Collapse
Affiliation(s)
- Aurélie Goyenvalle
- MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK.
| | | | | | | | | | | |
Collapse
|
41
|
Anthony K, Cirak S, Torelli S, Tasca G, Feng L, Arechavala-Gomeza V, Armaroli A, Guglieri M, Straathof CS, Verschuuren JJ, Aartsma-Rus A, Helderman-van den Enden P, Bushby K, Straub V, Sewry C, Ferlini A, Ricci E, Morgan JE, Muntoni F. Dystrophin quantification and clinical correlations in Becker muscular dystrophy: implications for clinical trials. ACTA ACUST UNITED AC 2011; 134:3547-59. [PMID: 22102647 DOI: 10.1093/brain/awr291] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy is caused by mutations in the DMD gene that disrupt the open reading frame and prevent the full translation of its protein product, dystrophin. Restoration of the open reading frame and dystrophin production can be achieved by exon skipping using antisense oligonucleotides targeted to splicing elements. This approach aims to transform the Duchenne muscular dystrophy phenotype to that of the milder disorder, Becker muscular dystrophy, typically caused by in-frame dystrophin deletions that allow the production of an internally deleted but partially functional dystrophin. There is ongoing debate regarding the functional properties of the different internally deleted dystrophins produced by exon skipping for different mutations; more insight would be valuable to improve and better predict the outcome of exon skipping clinical trials. To this end, we have characterized the clinical phenotype of 17 patients with Becker muscular dystrophy harbouring in-frame deletions relevant to on-going or planned exon skipping clinical trials for Duchenne muscular dystrophy and correlated it to the levels of dystrophin, and dystrophin-associated protein expression. The cohort of 17 patients, selected exclusively on the basis of their genotype, included 4 asymptomatic, 12 mild and 1 severe patient. All patients had dystrophin levels of >40% of control and significantly higher dystrophin (P = 0.013), β-dystroglycan (P = 0.025) and neuronal nitric oxide synthase (P = 0.034) expression was observed in asymptomatic individuals versus symptomatic patients with Becker muscular dystrophy. Furthermore, grouping the patients by deletion, patients with Becker muscular dystrophy with deletions with an end-point of exon 51 (the skipping of which could rescue the largest group of Duchenne muscular dystrophy deletions) showed significantly higher dystrophin levels (P = 0.034) than those with deletions ending with exon 53. This is the first quantitative study on both dystrophin and dystrophin-associated protein expression in patients with Becker muscular dystrophy with deletions relevant for on-going exon skipping trials in Duchenne muscular dystrophy. Taken together, our results indicate that all varieties of internally deleted dystrophin assessed in this study have the functional capability to provide a substantial clinical benefit to patients with Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Karen Anthony
- The Dubowitz Neuromuscular Centre, UCL, Institute of Child Health, London WC1N 1EH, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
van Putten M, Aartsma-Rus A. Opportunities and challenges for the development of antisense treatment in neuromuscular disorders. Expert Opin Biol Ther 2011; 11:1025-37. [PMID: 21510827 DOI: 10.1517/14712598.2011.579098] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Neuromuscular disorders are diseases of the musculature and/or the nervous system, generally leading to loss of muscle function. They are a frequent cause of disability and treatment options are often only symptomatic. Interestingly, for a number of neuromuscular disorders the application of antisense oligonucleotides has therapeutic potential. AREAS COVERED The authors describe how this approach is exploited for different neuromuscular diseases, focusing on literature published in the past 10 years. For each disease the opportunities of this approach, the state of the art, and current challenges are described. EXPERT OPINION A lot of progress has been made in the development of antisense-mediated approaches during recent years and they may become clinically applicable in the near future.
Collapse
Affiliation(s)
- Maaike van Putten
- Leiden University Medical Center, Department of Human Genetics, The Netherlands
| | | |
Collapse
|
43
|
Abstract
A milestone of molecular medicine is the identification of dystrophin gene mutation as the cause of Duchenne muscular dystrophy (DMD). Over the last 2 decades, major advances in dystrophin biology and gene delivery technology have created an opportunity to treat DMD with gene therapy. Remarkable success has been achieved in treating dystrophic mice. Several gene therapy strategies, including plasmid transfer, exon skipping, and adeno-associated virus-mediated microdystrophin therapy, have entered clinical trials. However, therapeutic benefit has not been realized in DMD patients. Bridging the gap between mice and humans is no doubt the most pressing issue facing DMD gene therapy now. In contrast to mice, dystrophin-deficient dogs are genetically and phenotypically similar to human patients. Preliminary gene therapy studies in the canine model may offer critical insights that cannot be obtained from murine studies. It is clear that the canine DMD model may represent an important link between mice and humans. Unfortunately, our current knowledge of dystrophic dogs is limited, and the full picture of disease progression remains to be clearly defined. We also lack rigorous outcome measures (such as in situ force measurement) to monitor therapeutic efficacy in dystrophic dogs. Undoubtedly, maintaining a dystrophic dog colony is technically demanding, and the cost of dog studies cannot be underestimated. A carefully coordinated effort from the entire DMD community is needed to make the best use of the precious dog resource. Successful DMD gene therapy may depend on valid translational studies in dystrophin-deficient dogs.
Collapse
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
44
|
Kyriakides T, Angelini C, Schaefer J, Sacconi S, Siciliano G, Vilchez JJ, Hilton-Jones D. EFNS guidelines on the diagnostic approach to pauci- or asymptomatic hyperCKemia. Eur J Neurol 2010; 17:767-73. [DOI: 10.1111/j.1468-1331.2010.03012.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Helderman-van den Enden ATJM, Straathof CSM, Aartsma-Rus A, den Dunnen JT, Verbist BM, Bakker E, Verschuuren JJGM, Ginjaar HB. Becker muscular dystrophy patients with deletions around exon 51; a promising outlook for exon skipping therapy in Duchenne patients. Neuromuscul Disord 2010; 20:251-4. [PMID: 20153965 DOI: 10.1016/j.nmd.2010.01.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2009] [Revised: 12/21/2009] [Accepted: 01/26/2010] [Indexed: 11/25/2022]
Abstract
Theoretically, 13% of patients with Duchenne muscular dystrophy may benefit from antisense-mediated skipping of exon 51 to restore the reading frame, which results in the production of a shortened dystrophin protein. We give a detailed description with longitudinal follow up of three patients with Becker muscular dystrophy with in-frame deletions in the DMD gene encompassing exon 51. Their internally deleted, but essentially functional, dystrophins are identical to those that are expected as end products in DMD patients treated with the exon 51 skipping therapy. The mild phenotype encourages further development of exon 51 skipping therapy.
Collapse
Affiliation(s)
- A T J M Helderman-van den Enden
- Center for Human and Clinical Genetics, The Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW Duchenne muscular dystrophy is a progressive muscle degenerative disease caused by dystrophin mutations. The purpose of this review is to highlight two emerging therapies designed to repair the primary genetic defect, called 'exon skipping' and 'nonsense codon suppression'. RECENT FINDINGS A drug, PTC124, was identified that suppresses nonsense codon translation termination. PTC124 can lead to restoration of some dystrophin expression in human Duchenne muscular dystrophy muscles with mutations resulting in premature stops. Two drugs developed for exon skipping, PRO051 and AVI-4658, result in the exclusion of exon 51 from mature mRNA. They can restore the translational reading frame to dystrophin transcripts from patients with a particular subset of dystrophin gene deletions and lead to some restoration of dystrophin expression in affected boys' muscle in vivo. Both approaches have concluded phase I trials with no serious adverse events. SUMMARY These novel therapies that act to correct the primary genetic defect of dystrophin deficiency are among the first generation of therapies tailored to correct specific mutations in humans. Thus, they represent paradigm forming approaches to personalized medicine with the potential to lead to life changing treatment for those affected by Duchenne muscular dystrophy.
Collapse
|