1
|
Martin C, Servais L. X-linked myotubular myopathy: an untreated treatable disease. Expert Opin Biol Ther 2025; 25:379-394. [PMID: 40042390 DOI: 10.1080/14712598.2025.2473430] [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: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION X-linked myotubular myopathy (XLMTM) is a life-threatening congenital disorder characterized by severe respiratory and motor impairment. This disease presents significant therapeutic challenges, with various strategies being explored to address its underlying pathology. Among these approaches, gene replacement therapy has demonstrated substantial functional improvements in clinical trials. However, safety issues emerged across different therapeutic approaches, highlighting the need for further research. AREAS COVERED This review provides a comprehensive analysis of the data gathered from natural history studies, preclinical models and clinical trials, with a particular focus on gene replacement therapy for XLMTM. The different therapeutic strategies are addressed, including their outcomes and associated safety concerns. EXPERT OPINION Despite the encouraging potential of gene therapy for XLMTM, the occurrence of safety challenges emphasizes the urgent need for a more comprehensive understanding of the disease's complex phenotype. Enhancing preclinical models to more accurately mimic the full spectrum of disease manifestations will be crucial for optimizing therapeutic strategies and reducing risks in future clinical applications.
Collapse
Affiliation(s)
- Cristina Martin
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Laurent Servais
- Department of Paediatrics, MDUK Oxford Neuromuscular Centre & NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Pediatrics, Neuromuscular Reference Center, University and University Hospital of Liège, Liège, Belgium
| |
Collapse
|
2
|
Mehmood H, Kasher PR, Barrett-Jolley R, Walmsley GL. Aligning with the 3Rs: alternative models for research into muscle development and inherited myopathies. BMC Vet Res 2024; 20:477. [PMID: 39425123 PMCID: PMC11488271 DOI: 10.1186/s12917-024-04309-z] [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: 03/15/2024] [Accepted: 09/30/2024] [Indexed: 10/21/2024] Open
Abstract
Inherited and acquired muscle diseases are an important cause of morbidity and mortality in human medical and veterinary patients. Researchers use models to study skeletal muscle development and pathology, improve our understanding of disease pathogenesis and explore new treatment options. Experiments on laboratory animals, including murine and canine models, have led to huge advances in congenital myopathy and muscular dystrophy research that have translated into clinical treatment trials in human patients with these debilitating and often fatal conditions. Whilst animal experimentation has enabled many significant and impactful discoveries that otherwise may not have been possible, we have an ethical and moral, and in many countries also a legal, obligation to consider alternatives. This review discusses the models available as alternatives to mammals for muscle development, biology and disease research with a focus on inherited myopathies. Cell culture models can be used to replace animals for some applications: traditional monolayer cultures (for example, using the immortalised C2C12 cell line) are accessible, tractable and inexpensive but developmentally limited to immature myotube stages; more recently, developments in tissue engineering have led to three-dimensional cultures with improved differentiation capabilities. Advances in computer modelling and an improved understanding of pathogenetic mechanisms are likely to herald new models and opportunities for replacement. Where this is not possible, a 3Rs approach advocates partial replacement with the use of less sentient animals (including invertebrates (such as worms Caenorhabditis elegans and fruit flies Drosophila melanogaster) and embryonic stages of small vertebrates such as the zebrafish Danio rerio) alongside refinement of experimental design and improved research practices to reduce the numbers of animals used and the severity of their experience. An understanding of the advantages and disadvantages of potential models is essential for researchers to determine which can best facilitate answering a specific scientific question. Applying 3Rs principles to research not only improves animal welfare but generates high-quality, reproducible and reliable data with translational relevance to human and animal patients.
Collapse
Affiliation(s)
- Hashir Mehmood
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Allianceand the, University of Manchester , Manchester, M6 8HD, UK
| | - Richard Barrett-Jolley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Gemma L Walmsley
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, Faculty of Health and Lifesciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
- Department of Small Animal Clinical Sciences, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, South Wirral, Neston, CH64 7TE, UK.
| |
Collapse
|
3
|
Dai N, Groenendyk J, Michalak M. Interplay between myotubularins and Ca 2+ homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119739. [PMID: 38710289 DOI: 10.1016/j.bbamcr.2024.119739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024]
Abstract
The myotubularin family, encompassing myotubularin 1 (MTM1) and 14 myotubularin-related proteins (MTMRs), represents a conserved group of phosphatases featuring a protein tyrosine phosphatase domain. Nine members are characterized by an active phosphatase domain C(X)5R, dephosphorylating the D3 position of PtdIns(3)P and PtdIns(3,5)P2. Mutations in myotubularin genes result in human myopathies, and several neuropathies including X-linked myotubular myopathy and Charcot-Marie-Tooth type 4B. MTM1, MTMR6 and MTMR14 also contribute to Ca2+ signaling and Ca2+ homeostasis that play a key role in many MTM-dependent myopathies and neuropathies. Here we explore the evolving roles of MTM1/MTMRs, unveiling their influence on critical aspects of Ca2+ signaling pathways.
Collapse
Affiliation(s)
- Ning Dai
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta T6G 2H7, Canada.
| |
Collapse
|
4
|
Giraud Q, Spiegelhalter C, Messaddeq N, Laporte J. MTM1 overexpression prevents and reverts BIN1-related centronuclear myopathy. Brain 2023; 146:4158-4173. [PMID: 37490306 PMCID: PMC10545525 DOI: 10.1093/brain/awad251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023] Open
Abstract
Centronuclear and myotubular myopathies (CNM) are rare and severe genetic diseases associated with muscle weakness and atrophy as well as intracellular disorganization of myofibres. The main mutated proteins control lipid and membrane dynamics and are the lipid phosphatase myotubularin (MTM1), and the membrane remodelling proteins amphiphysin 2 (BIN1) and dynamin 2 (DNM2). There is no available therapy. Here, to validate a novel therapeutic strategy for BIN1- and DNM2-CNM, we evaluated adeno-associated virus-mediated MTM1 (AAV-MTM1 ) overexpression in relevant mouse models. Early systemic MTM1 overexpression prevented the development of the CNM pathology in Bin1mck-/- mice, while late intramuscular MTM1 expression partially reverted the established phenotypes after only 4 weeks of treatment. However, AAV-MTM1 injection did not change the DNM2-CNM mouse phenotypes. We investigated the mechanism of the rescue of the myopathy in BIN1-CNM and found that the lipid phosphatase activity of MTM1 was essential for the rescue of muscle atrophy and myofibre hypotrophy but dispensable for the rescue of myofibre disorganization including organelle mis-position and T-tubule defects. Furthermore, the improvement of T-tubule organization correlated with normalization of key regulators of T-tubule morphogenesis, dysferlin and caveolin. Overall, these data support the inclusion of BIN1-CNM patients in an AAV-MTM1 clinical trial.
Collapse
Affiliation(s)
- Quentin Giraud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Coralie Spiegelhalter
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Nadia Messaddeq
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404, Illkirch, France
| |
Collapse
|
5
|
Moura E, Tasqueti UI, Mangrich-Rocha RMV, Filho JRE, de Farias MR, Pimpão CT. Inborn Errors of Metabolism in Dogs: Historical, Metabolic, Genetic, and Clinical Aspects. Top Companion Anim Med 2022; 51:100731. [DOI: 10.1016/j.tcam.2022.100731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
|
6
|
Kopke MA, Diane Shelton G, Lyons LA, Wall MJ, Pemberton S, Gedye KR, Owen R, Guo LT, Buckley RM, Valencia JA, Jones BR. X-linked myotubular myopathy associated with an MTM1 variant in a Maine coon cat. J Vet Intern Med 2022; 36:1800-1805. [PMID: 35962713 PMCID: PMC9511081 DOI: 10.1111/jvim.16509] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/19/2022] [Indexed: 12/05/2022] Open
Abstract
Objective Describe the clinical course and diagnostic and genetic findings in a cat with X‐linked myotubular myopathy. Case Summary A 7‐month‐old male Maine coon was evaluated for progressively worsening gait abnormalities and generalized weakness. Neurolocalization was to the neuromuscular system. Genetic testing for spinal muscular atrophy (LIX1) was negative. Given the progressive nature and suspected poor long‐term prognosis, the owners elected euthanasia. Histopathology of skeletal muscle obtained post‐mortem disclosed numerous rounded atrophic or hypotrophic fibers with internal nuclei or central basophilic staining. Using oxidative reactions mediated by cytochrome C oxidase and succinic dehydrogenase, scattered myofibers were observed to have central dark staining structures and a “ring‐like” appearance. Given the cat's age and clinical history, a congenital myopathy was considered most likely, with the central nuclei and “ring‐like” changes consistent with either centronuclear or myotubular myopathy. Whole genome sequencing identified an underlying missense variant in myotubularin 1 (MTM1), a known candidate gene for X‐linked myotubular myopathy. New or Unique Information Provided This case is the first report of X‐linked myotubular myopathy in a cat with an MTM1 missense mutation. Maine coon cat breeders may consider screening for this variant to prevent production of affected cats and to eradicate the variant from the breeding population.
Collapse
Affiliation(s)
- Matthew A Kopke
- School of Veterinary Science, Massey University, Palmerston North, New Zealand.,Veterinary Nutrition Group, Le Fel, France
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Leslie A Lyons
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Meredith J Wall
- School of Veterinary Science, Massey University, Palmerston North, New Zealand.,Veterinary Nutrition Group, Le Fel, France
| | - Sarah Pemberton
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Kristene R Gedye
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Rebecca Owen
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Reuben M Buckley
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Juan A Valencia
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | | | - Boyd R Jones
- School of Veterinary Science, Massey University, Palmerston North, New Zealand
| |
Collapse
|
7
|
Shelton GD, Minor KM, Guo LT, Thomas-Hollands A, Walsh KA, Friedenberg SG, Cullen JN, Mickelson JR. An EHPB1L1 Nonsense Mutation Associated with Congenital Dyserythropoietic Anemia and Polymyopathy in Labrador Retriever Littermates. Genes (Basel) 2022; 13:1427. [PMID: 36011338 PMCID: PMC9407898 DOI: 10.3390/genes13081427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/30/2022] Open
Abstract
In this report, we describe a novel genetic basis for congenital dyserythropoietic anemia and polymyopathy in Labrador Retriever littermates characterized by incidental detection of marked microcytosis, inappropriate metarubricytosis, pelvic limb weakness and muscle atrophy. A similar syndrome has been described in English Springer Spaniel littermates with an early onset of anemia, megaesophagus, generalized muscle atrophy and cardiomyopathy. Muscle histopathology in both breeds showed distinctive pathological changes consistent with congenital polymyopathy. Using whole genome sequencing and mapping to the CanFam4 (Canis lupus familiaris reference assembly 4), a nonsense variant in the EHBP1L1 gene was identified in a homozygous form in the Labrador Retriever littermates. The mutation produces a premature stop codon that deletes approximately 90% of the protein. This variant was not present in the English Springer Spaniels. Currently, EHPB1L1 is described as critical to actin cytoskeletal organization and apical-directed transport in polarized epithelial cells, and through connections with Rab8 and a BIN1-dynamin complex generates membrane vesicles in the endocytic recycling compartment. Furthermore, EHBP1L1 knockout mice die early and develop severe anemia. The connection of EHBP1L1 to BIN1 and DMN2 functions is particularly interesting due to BIN1 and DMN2 mutations being causative in forms of centronuclear myopathy. This report, along with an independent study conducted by another group, are the first reports of an association of EHBP1L1 mutations with congenital dyserythropoietic anemia and polymyopathy.
Collapse
Affiliation(s)
- G. Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Katie M. Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Ling T. Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Alison Thomas-Hollands
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Koranda A. Walsh
- Department of Clinical Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Jonah N. Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - James R. Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
8
|
Sarikaya E, Sabha N, Volpatti J, Pannia E, Maani N, Gonorazky HD, Celik A, Liang Y, Onofre-Oliveira P, Dowling JJ. Natural history of a mouse model of X-linked myotubular myopathy. Dis Model Mech 2022; 15:276037. [PMID: 35694952 PMCID: PMC9346535 DOI: 10.1242/dmm.049342] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 06/06/2022] [Indexed: 11/20/2022] Open
Abstract
X-linked myotubular myopathy (XLMTM) is a severe monogenetic disorder of the skeletal muscle. It is caused by loss-of-expression/function mutations in the myotubularin (MTM1) gene. Much of what is known about the disease, as well as the treatment strategies, has been uncovered through experimentation in pre-clinical models, particularly the Mtm1 gene knockout mouse line (Mtm1 KO). Despite this understanding, and the identification of potential therapies, much remains to be understood about XLMTM disease pathomechanisms, and about the normal functions of MTM1 in muscle development. To lay the groundwork for addressing these knowledge gaps, we performed a natural history study of Mtm1 KO mice. This included longitudinal comparative analyses of motor phenotype, transcriptome and proteome profiles, muscle structure and targeted molecular pathways. We identified age-associated changes in gene expression, mitochondrial function, myofiber size and key molecular markers, including DNM2. Importantly, some molecular and histopathologic changes preceded overt phenotypic changes, while others, such as triad structural alternations, occurred coincidentally with the presence of severe weakness. In total, this study provides a comprehensive longitudinal evaluation of the murine XLMTM disease process, and thus provides a critical framework for future investigations. Summary: This study provides a comprehensive and longitudinal molecular and phenotypic evaluation of the disease process of X-linked myotubular myopathy (XLMTM) in a murine model.
Collapse
Affiliation(s)
- Ege Sarikaya
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Nesrin Sabha
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada
| | - Jonathan Volpatti
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Emanuela Pannia
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Nika Maani
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Hernan D Gonorazky
- Division of Neurology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada
| | - Alper Celik
- Centre for Computational Medicine, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada
| | - Yijng Liang
- Centre for Computational Medicine, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada
| | - Paula Onofre-Oliveira
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Molecular Genetics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.,Division of Neurology, Hospital for Sick Children, 686 Bay Street, Toronto, Ontario, M5G 1X8, Canada.,Departments of Paediatrics, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| |
Collapse
|
9
|
Böhm J, Barthélémy I, Landwerlin C, Blanchard-Gutton N, Relaix F, Blot S, Laporte J, Tiret L. A dog model for centronuclear myopathy carrying the most common DNM2 mutation. Dis Model Mech 2022; 15:274622. [PMID: 35244154 PMCID: PMC9016898 DOI: 10.1242/dmm.049219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/21/2022] [Indexed: 12/17/2022] Open
Abstract
Mutations in DNM2 cause autosomal dominant centronuclear myopathy (ADCNM), a rare disease characterized by skeletal muscle weakness and structural anomalies of the myofibres, including nuclear centralization and mitochondrial mispositioning. Following the clinical report of a Border Collie male with exercise intolerance and histopathological hallmarks of CNM on the muscle biopsy, we identified the c.1393C>T (R465W) mutation in DNM2, corresponding to the most common ADCNM mutation in humans. In order to establish a large animal model for longitudinal and preclinical studies on the muscle disorder, we collected sperm samples from the Border Collie male and generated a dog cohort for subsequent clinical, genetic and histological investigations. Four of the five offspring carried the DNM2 mutation and showed muscle atrophy and a mildly impaired gait. Morphological examinations of transverse muscle sections revealed CNM-typical fibres with centralized nuclei and remodelling of the mitochondrial network. Overall, the DNM2-CNM dog represents a faithful animal model for the human disorder, allows the investigation of ADCNM disease progression, and constitutes a valuable complementary tool to validate innovative therapies established in mice.
Collapse
Affiliation(s)
- Johann Böhm
- Médecine translationnelle et neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U 1258, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Inès Barthélémy
- Neuroscience et psychiatrie, Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France.,Ecole nationale vétérinaire d'Alfort, IMRB, 94700 Maisons-Alfort, France.,EFS, IMRB, 94017 Créteil Cedex, France
| | - Charlène Landwerlin
- Médecine translationnelle et neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U 1258, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Nicolas Blanchard-Gutton
- Neuroscience et psychiatrie, Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France.,Ecole nationale vétérinaire d'Alfort, IMRB, 94700 Maisons-Alfort, France.,EFS, IMRB, 94017 Créteil Cedex, France
| | - Frédéric Relaix
- Neuroscience et psychiatrie, Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France.,Ecole nationale vétérinaire d'Alfort, IMRB, 94700 Maisons-Alfort, France.,EFS, IMRB, 94017 Créteil Cedex, France
| | - Stéphane Blot
- Neuroscience et psychiatrie, Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France.,Ecole nationale vétérinaire d'Alfort, IMRB, 94700 Maisons-Alfort, France.,EFS, IMRB, 94017 Créteil Cedex, France
| | - Jocelyn Laporte
- Médecine translationnelle et neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U 1258, CNRS UMR 7104, Université de Strasbourg, 67404 Illkirch, France
| | - Laurent Tiret
- Neuroscience et psychiatrie, Université Paris-Est Créteil, INSERM, IMRB, 94010 Créteil, France.,Ecole nationale vétérinaire d'Alfort, IMRB, 94700 Maisons-Alfort, France.,EFS, IMRB, 94017 Créteil Cedex, France
| |
Collapse
|
10
|
Lawlor MW, Dowling JJ. X-linked myotubular myopathy. Neuromuscul Disord 2021; 31:1004-1012. [PMID: 34736623 DOI: 10.1016/j.nmd.2021.08.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/28/2022]
Abstract
X-linked myotubular myopathy (XLMTM) is a severe congenital muscle disease caused by mutation in the MTM1 gene. MTM1 encodes myotubularin (MTM1), an endosomal phosphatase that acts to dephosphorylate key second messenger lipids PI3P and PI3,5P2. XLMTM is clinically characterized by profound muscle weakness and associated with multiple disabilities (including ventilator and wheelchair dependence) and early death in most affected individuals. The disease is classically defined by characteristic changes observed on muscle biopsy, including centrally located nuclei, myofiber hypotrophy, and organelle disorganization. In this review, we highlight the clinical and pathologic features of the disease, present concepts related to disease pathomechanisms, and present recent advances in therapy development.
Collapse
Affiliation(s)
- Michael W Lawlor
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - James J Dowling
- Division of Neurology and Program for Genetics and Genome Biology, Hospital for Sick Children, 555 University Ave., Toronto, ON M5G 1X8, Canada; Departments of Paediatrics and Molecular Genetics, University of Toronto, Canada.
| |
Collapse
|
11
|
Shelton GD, Minor KM, Guo LT, Friedenberg SG, Cullen JN, Hord JM, Venzke D, Anderson ME, Devereaux M, Prouty SJ, Handelman C, Campbell KP, Mickelson JR. Muscular dystrophy-dystroglycanopathy in a family of Labrador retrievers with a LARGE1 mutation. Neuromuscul Disord 2021; 31:1169-1178. [PMID: 34654610 PMCID: PMC8963908 DOI: 10.1016/j.nmd.2021.07.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
Alpha-dystroglycan (αDG) is a highly glycosylated cell surface protein with a significant role in cell-to-extracellular matrix interactions in muscle. αDG interaction with extracellular ligands relies on the activity of the LARGE1 glycosyltransferase that synthesizes and extends the heteropolysaccharide matriglycan. Abnormalities in αDG glycosylation and formation of matriglycan are the pathogenic mechanisms for the dystroglycanopathies, a group of congenital muscular dystrophies. Muscle biopsies were evaluated from related 6-week-old Labrador retriever puppies with poor suckling, small stature compared to normal litter mates, bow-legged stance and markedly elevated creatine kinase activities. A dystrophic phenotype with marked degeneration and regeneration, multifocal mononuclear cell infiltration and endomysial fibrosis was identified on muscle cryosections. Single nucleotide polymorphism (SNP) array genotyping data on the family members identified three regions of homozygosity in 4 cases relative to 8 controls. Analysis of whole genome sequence data from one of the cases identified a stop codon mutation in the LARGE1 gene that truncates 40% of the protein. Immunofluorescent staining and western blotting demonstrated the absence of matriglycan in skeletal muscle and heart from affected dogs. Compared to control, LARGE enzyme activity was not detected. This is the first report of a dystroglycanopathy in dogs.
Collapse
Affiliation(s)
- G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709 United States.
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709 United States
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Jonah N Cullen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| | - Jeffrey M Hord
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - David Venzke
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Mary E Anderson
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Megan Devereaux
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Sally J Prouty
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - Caryl Handelman
- Veterinary Housecalls of Long Island, Commack, NY 11725 United States
| | - Kevin P Campbell
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, Department of Molecular Physiology and Biophysics and Department of Neurology, Howard Hughes Medical Institute, Roy J and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA 52242 United States
| | - James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108 United States
| |
Collapse
|
12
|
Gómez-Oca R, Cowling BS, Laporte J. Common Pathogenic Mechanisms in Centronuclear and Myotubular Myopathies and Latest Treatment Advances. Int J Mol Sci 2021; 22:11377. [PMID: 34768808 PMCID: PMC8583656 DOI: 10.3390/ijms222111377] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/18/2023] Open
Abstract
Centronuclear myopathies (CNM) are rare congenital disorders characterized by muscle weakness and structural defects including fiber hypotrophy and organelle mispositioning. The main CNM forms are caused by mutations in: the MTM1 gene encoding the phosphoinositide phosphatase myotubularin (myotubular myopathy), the DNM2 gene encoding the mechanoenzyme dynamin 2, the BIN1 gene encoding the membrane curvature sensing amphiphysin 2, and the RYR1 gene encoding the skeletal muscle calcium release channel/ryanodine receptor. MTM1, BIN1, and DNM2 proteins are involved in membrane remodeling and trafficking, while RyR1 directly regulates excitation-contraction coupling (ECC). Several CNM animal models have been generated or identified, which confirm shared pathological anomalies in T-tubule remodeling, ECC, organelle mispositioning, protein homeostasis, neuromuscular junction, and muscle regeneration. Dynamin 2 plays a crucial role in CNM physiopathology and has been validated as a common therapeutic target for three CNM forms. Indeed, the promising results in preclinical models set up the basis for ongoing clinical trials. Another two clinical trials to treat myotubular myopathy by MTM1 gene therapy or tamoxifen repurposing are also ongoing. Here, we review the contribution of the different CNM models to understanding physiopathology and therapy development with a focus on the commonly dysregulated pathways and current therapeutic targets.
Collapse
Affiliation(s)
- Raquel Gómez-Oca
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
- Dynacure, 67400 Illkirch, France;
| | | | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, 67400 Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, 67400 Illkirch, France
- Strasbourg University, 67081 Strasbourg, France
| |
Collapse
|
13
|
Djeddi S, Reiss D, Menuet A, Freismuth S, de Carvalho Neves J, Djerroud S, Massana-Muñoz X, Sosson AS, Kretz C, Raffelsberger W, Keime C, Dorchies OM, Thompson J, Laporte J. Multi-omics comparisons of different forms of centronuclear myopathies and the effects of several therapeutic strategies. Mol Ther 2021; 29:2514-2534. [PMID: 33940157 DOI: 10.1016/j.ymthe.2021.04.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
Omics analyses are powerful methods to obtain an integrated view of complex biological processes, disease progression, or therapy efficiency. However, few studies have compared different disease forms and different therapy strategies to define the common molecular signatures representing the most significant implicated pathways. In this study, we used RNA sequencing and mass spectrometry to profile the transcriptomes and proteomes of mouse models for three forms of centronuclear myopathies (CNMs), untreated or treated with either a drug (tamoxifen), antisense oligonucleotides reducing the level of dynamin 2 (DNM2), or following modulation of DNM2 or amphiphysin 2 (BIN1) through genetic crosses. Unsupervised analysis and differential gene and protein expression were performed to retrieve CNM molecular signatures. Longitudinal studies before, at, and after disease onset highlighted potential disease causes and consequences. Main pathways in the common CNM disease signature include muscle contraction, regeneration and inflammation. The common therapy signature revealed novel potential therapeutic targets, including the calcium regulator sarcolipin. We identified several novel biomarkers validated in muscle and/or plasma through RNA quantification, western blotting, and enzyme-linked immunosorbent assay (ELISA) assays, including ANXA2 and IGFBP2. This study validates the concept of using multi-omics approaches to identify molecular signatures common to different disease forms and therapeutic strategies.
Collapse
Affiliation(s)
- Sarah Djeddi
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Alexia Menuet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Sébastien Freismuth
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Juliana de Carvalho Neves
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Sarah Djerroud
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Xènia Massana-Muñoz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Anne-Sophie Sosson
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Christine Kretz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Wolfgang Raffelsberger
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Céline Keime
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France
| | - Olivier M Dorchies
- Pharmaceutical Biochemistry, Institute of Pharmaceutical Sciences of Western Switzerland (ISPSO), University of Geneva, 1211 Geneva, Switzerland
| | - Julie Thompson
- Complex Systems and Translational Bioinformatics (CSTB), ICube Laboratory-CNRS, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, 67000 Strasbourg, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), CNRS UMR7104, INSERM U1258, Université de Strasbourg, 67404 Illkirch, France.
| |
Collapse
|
14
|
Pegram C, Woolley C, Brodbelt DC, Church DB, O'Neill DG. Disorder predispositions and protections of Labrador Retrievers in the UK. Sci Rep 2021; 11:13988. [PMID: 34262062 PMCID: PMC8280121 DOI: 10.1038/s41598-021-93379-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 06/21/2021] [Indexed: 02/06/2023] Open
Abstract
The Labrador Retriever is one of the most popular dog breeds worldwide, therefore it is important to have reliable evidence on the general health issues of the breed. Using anonymised veterinary clinical data from the VetCompass Programme, this study aimed to explore the relative risk to common disorders in the Labrador Retriever. The clinical records of a random sample of dogs were reviewed to extract the most definitive diagnoses for all disorders recorded during 2016. A list of disorders was generated, including the 30 most common disorders in Labrador Retrievers and the 30 most common disorders in non-Labrador Retrievers. Multivariable logistic regression was used to report the odds of each of these disorders in 1462 (6.6%) Labrador Retrievers compared with 20,786 (93.4%) non-Labrador Retrievers. At a specific-level of diagnostic precision, after accounting for confounding, Labrador Retrievers had significantly increased odds of 12/35 (34.3%) disorders compared to non-Labrador Retrievers; osteoarthritis (OR 2.83) had the highest odds. Conversely, Labrador Retrievers had reduced odds of 7/35 (20.0%) disorders; patellar luxation (OR 0.18) had the lowest odds. This study provides useful information about breed-specific disorder predispositions and protections, which future research could evaluate further to produce definitive guidance for Labrador Retriever breeders and owners.
Collapse
Affiliation(s)
- Camilla Pegram
- Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK.
| | - Charlotte Woolley
- The Roslin Institute and the Royal (Dick), School of Veterinary Studies, The University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, UK
| | - Dave C Brodbelt
- Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - David B Church
- Clinical Sciences and Services, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| | - Dan G O'Neill
- Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, AL9 7TA, Herts, UK
| |
Collapse
|
15
|
Thomas-Hollands A, Shelton GD, Guo LT, Loughran K, Kaiman G, A Hutton T, Walsh KA. Congenital dyserythropoiesis and polymyopathy without cardiac disease in male Labrador retriever littermates. J Vet Intern Med 2021; 35:2409-2414. [PMID: 34227150 PMCID: PMC8478025 DOI: 10.1111/jvim.16214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 11/30/2022] Open
Abstract
Background Two Labrador retriever littermates were identified based on incidentally noted marked microcytosis and inappropriate metarubricytosis. Muscle atrophy was noted and associated with distinctive pathological findings in biopsy samples from 1 dog studied. The disorder represents a rare clinical entity of suspected congenital dyserythropoiesis and polymyopathy. Clinicopathologic changes were similar to a previously reported syndrome of congenital dyserythropoiesis, congenital polymyopathy, and cardiac disease in 3 related English Springer Spaniel (ESS) dogs, but the dogs reported here did not have apparent cardiac disease. Interventions Bone marrow aspiration, electromyography, muscle biopsies, and an echocardiogram were performed on dog 1. Results supported dyserythropoiesis and congenital polymyopathy similar to reports in ESS dogs, but did not identify obvious cardiac disease. Conclusion The clinicopathologic changes of dyserythropoiesis and polymyopathy provide an easily recognizable phenotype for what appears to be a low morbidity syndrome. Early recognition may decrease unnecessary testing or euthanasia.
Collapse
Affiliation(s)
- Alison Thomas-Hollands
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - G Diane Shelton
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Ling T Guo
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Kerry Loughran
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory Kaiman
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tabitha A Hutton
- Metropolitan Veterinary Associates, Norristown, Pennsylvania, USA
| | - Koranda A Walsh
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
16
|
Luo S, Li Q, Lin J, Murphy Q, Marty I, Zhang Y, Kazerounian S, Agrawal PB. SPEG binds with desmin and its deficiency causes defects in triad and focal adhesion proteins. Hum Mol Genet 2020; 29:3882-3891. [PMID: 33355670 DOI: 10.1093/hmg/ddaa276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/17/2020] [Accepted: 12/09/2020] [Indexed: 11/13/2022] Open
Abstract
Striated preferentially expressed gene (SPEG), a member of the myosin light chain kinase family, is localized at the level of triad surrounding myofibrils in skeletal muscles. In humans, SPEG mutations are associated with centronuclear myopathy and cardiomyopathy. Using a striated muscle-specific Speg-knockout (KO) mouse model, we have previously shown that SPEG is critical for triad maintenance and calcium handling. Here, we further examined the molecular function of SPEG and characterized the effects of SPEG deficiency on triad and focal adhesion proteins. We used yeast two-hybrid assay, and identified desmin, an intermediate filament protein, to interact with SPEG and confirmed this interaction by co-immunoprecipitation. Using domain-mapping assay, we defined that Ig-like and fibronectin III domains of SPEG interact with rod domain of desmin. In skeletal muscles, SPEG depletion leads to desmin aggregates in vivo and a shift in desmin equilibrium from soluble to insoluble fraction. We also profiled the expression and localization of triadic proteins in Speg-KO mice using western blot and immunofluorescence. The amount of RyR1 and triadin were markedly reduced, whereas DHPRα1, SERCA1 and triadin were abnormally accumulated in discrete areas of Speg-KO myofibers. In addition, Speg-KO muscles exhibited internalized vinculin and β1 integrin, both of which are critical components of the focal adhesion complex. Further, β1 integrin was abnormally accumulated in early endosomes of Speg-KO myofibers. These results demonstrate that SPEG-deficient skeletal muscles exhibit several pathological features similar to those seen in MTM1 deficiency. Defects of shared cellular pathways may underlie these structural and functional abnormalities in both types of diseases.
Collapse
Affiliation(s)
- Shiyu Luo
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qifei Li
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jasmine Lin
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Quinn Murphy
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Isabelle Marty
- Grenoble Institut Neurosciences, Inserm, U1216, University Grenoble Alpes, 38000 Grenoble, France
| | - Yuanfan Zhang
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shideh Kazerounian
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pankaj B Agrawal
- Division of Newborn Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Buscara L, Gross DA, Daniele N. Of rAAV and Men: From Genetic Neuromuscular Disorder Efficacy and Toxicity Preclinical Studies to Clinical Trials and Back. J Pers Med 2020; 10:E258. [PMID: 33260623 PMCID: PMC7768510 DOI: 10.3390/jpm10040258] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Neuromuscular disorders are a large group of rare pathologies characterised by skeletal muscle atrophy and weakness, with the common involvement of respiratory and/or cardiac muscles. These diseases lead to life-long motor deficiencies and specific organ failures, and are, in their worst-case scenarios, life threatening. Amongst other causes, they can be genetically inherited through mutations in more than 500 different genes. In the last 20 years, specific pharmacological treatments have been approved for human usage. However, these "à-la-carte" therapies cover only a very small portion of the clinical needs and are often partially efficient in alleviating the symptoms of the disease, even less so in curing it. Recombinant adeno-associated virus vector-mediated gene transfer is a more general strategy that could be adapted for a large majority of these diseases and has proved very efficient in rescuing the symptoms in many neuropathological animal models. On this solid ground, several clinical trials are currently being conducted with the whole-body delivery of the therapeutic vectors. This review recapitulates the state-of-the-art tools for neuron and muscle-targeted gene therapy, and summarises the main findings of the spinal muscular atrophy (SMA), Duchenne muscular dystrophy (DMD) and X-linked myotubular myopathy (XLMTM) trials. Despite promising efficacy results, serious adverse events of various severities were observed in these trials. Possible leads for second-generation products are also discussed.
Collapse
Affiliation(s)
| | - David-Alexandre Gross
- Genethon, 91000 Evry, France; (L.B.); (D.-A.G.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | | |
Collapse
|
18
|
Ross JA, Tasfaout H, Levy Y, Morgan J, Cowling BS, Laporte J, Zanoteli E, Romero NB, Lowe DA, Jungbluth H, Lawlor MW, Mack DL, Ochala J. rAAV-related therapy fully rescues myonuclear and myofilament function in X-linked myotubular myopathy. Acta Neuropathol Commun 2020; 8:167. [PMID: 33076971 PMCID: PMC7574461 DOI: 10.1186/s40478-020-01048-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 10/02/2020] [Indexed: 01/17/2023] Open
Abstract
X-linked myotubular myopathy (XLMTM) is a life-threatening skeletal muscle disease caused by mutations in the MTM1 gene. XLMTM fibres display a population of nuclei mispositioned in the centre. In the present study, we aimed to explore whether positioning and overall distribution of nuclei affects cellular organization and contractile function, thereby contributing to muscle weakness in this disease. We also assessed whether gene therapy alters nuclear arrangement and function. We used tissue from human patients and animal models, including XLMTM dogs that had received increasing doses of recombinant AAV8 vector restoring MTM1 expression (rAAV8-cMTM1). We then used single isolated muscle fibres to analyze nuclear organization and contractile function. In addition to the expected mislocalization of nuclei in the centre of muscle fibres, a novel form of nuclear mispositioning was observed: irregular spacing between those located at the fibre periphery, and an overall increased number of nuclei, leading to dramatically smaller and inconsistent myonuclear domains. Nuclear mislocalization was associated with decreases in global nuclear synthetic activity, contractile protein content and intrinsic myofilament force production. A contractile deficit originating at the myofilaments, rather than mechanical interference by centrally positioned nuclei, was supported by experiments in regenerated mouse muscle. Systemic administration of rAAV8-cMTM1 at doses higher than 2.5 × 1013 vg kg−1 allowed a full rescue of all these cellular defects in XLMTM dogs. Altogether, these findings identify previously unrecognized pathological mechanisms in human and animal XLMTM, associated with myonuclear defects and contractile filament function. These defects can be reversed by gene therapy restoring MTM1 expression in dogs with XLMTM.
Collapse
|
19
|
Morales L, Gambhir Y, Bennett J, Stedman HH. Broader Implications of Progressive Liver Dysfunction and Lethal Sepsis in Two Boys following Systemic High-Dose AAV. Mol Ther 2020; 28:1753-1755. [PMID: 32710826 PMCID: PMC7363592 DOI: 10.1016/j.ymthe.2020.07.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Leon Morales
- Biomedical Graduate Studies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuva Gambhir
- School of Arts and Sciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean Bennett
- Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hansell H Stedman
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania and Corporal Michael Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
Bolduc V, Minor KM, Hu Y, Kaur R, Friedenberg SG, Van Buren S, Guo LT, Glennon JC, Marioni-Henry K, Mickelson JR, Bönnemann CG, Shelton GD. Pathogenic variants in COL6A3 cause Ullrich-like congenital muscular dystrophy in young Labrador Retriever dogs. Neuromuscul Disord 2020; 30:360-367. [PMID: 32439203 PMCID: PMC7292757 DOI: 10.1016/j.nmd.2020.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 01/22/2023]
Abstract
The collagen VI-related muscular dystrophies in people include a broad spectrum of diseases ranging from the severe Ullrich congenital muscular dystrophy to the mild Bethlem myopathy. Clinical features are attributable to both muscle and connective tissue and include progressive muscle weakness and respiratory failure, hyperlaxity of distal joints, and progressive contracture of large joints. Here we describe two different COL6A3 pathogenic variants in Labrador Retriever dogs that result in autosomal recessive or autosomal dominant congenital myopathies with hyperlaxity of distal joints and joint contracture, similar to the condition in people.
Collapse
Affiliation(s)
- Véronique Bolduc
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3705, USA
| | - Katie M Minor
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Ying Hu
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3705, USA
| | - Rupleen Kaur
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3705, USA
| | - Steven G Friedenberg
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Samantha Van Buren
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Ling T Guo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709, USA
| | | | - Katia Marioni-Henry
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, United Kingdom
| | - James R Mickelson
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN, USA
| | - Carsten G Bönnemann
- Neuromuscular and Neurogenetic Disorders of Childhood Section, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-3705, USA.
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA 92093-0709, USA.
| |
Collapse
|
21
|
Barthélémy I, Hitte C, Tiret L. The Dog Model in the Spotlight: Legacy of a Trustful Cooperation. J Neuromuscul Dis 2020; 6:421-451. [PMID: 31450509 PMCID: PMC6918919 DOI: 10.3233/jnd-190394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dogs have long been used as a biomedical model system and in particular as a preclinical proof of concept for innovative therapies before translation to humans. A recent example of the utility of this animal model is the promising myotubularin gene delivery in boys affected by X-linked centronuclear myopathy after successful systemic, long-term efficient gene therapy in Labrador retrievers. Mostly, this is due to unique features that make dogs an optimal system. The continuous emergence of spontaneous inherited disorders enables the identification of reliable complementary molecular models for human neuromuscular disorders (NMDs). Dogs’ characteristics including size, lifespan and unprecedented medical care level allow a comprehensive longitudinal description of diseases. Moreover, the highly similar pathogenic mechanisms with human patients yield to translational robustness. Finally, interindividual phenotypic heterogeneity between dogs helps identifying modifiers and anticipates precision medicine issues. This review article summarizes the present list of molecularly characterized dog models for NMDs and provides an exhaustive list of the clinical and paraclinical assays that have been developed. This toolbox offers scientists a sensitive and reliable system to thoroughly evaluate neuromuscular function, as well as efficiency and safety of innovative therapies targeting these NMDs. This review also contextualizes the model by highlighting its unique genetic value, shaped by the long-term coevolution of humans and domesticated dogs. Because the dog is one of the most protected research animal models, there is considerable opposition to include it in preclinical projects, posing a threat to the use of this model. We thus discuss ethical issues, emphasizing that unlike many other models, the dog also benefits from its contribution to comparative biomedical research with a drastic reduction in the prevalence of morbid alleles in the breeding stock and an improvement in medical care.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Christophe Hitte
- CNRS, University of Rennes 1, UMR 6290, IGDR, Faculty of Medicine, SFR Biosit, Rennes, France
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
22
|
A mutation in MTM1 causes X-Linked myotubular myopathy in Boykin spaniels. Neuromuscul Disord 2020; 30:353-359. [PMID: 32417001 DOI: 10.1016/j.nmd.2020.02.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/18/2020] [Accepted: 02/28/2020] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to report the findings of clinical and genetic evaluation of a 3-month old male Boykin spaniel (the proband) that presented with progressive weakness. The puppy underwent a physical and neurological examination, serum biochemistry and complete blood cell count, electrophysiological testing, muscle biopsy and whole genome sequencing. Clinical evaluation revealed generalized neuromuscular weakness with tetraparesis and difficulty holding the head up and a dropped jaw. There was diffuse spontaneous activity on electromyography, most severe in the cervical musculature. Nerve conduction studies were normal, the findings were interpreted as consistent with a myopathy. Skeletal muscle was grossly abnormal on biopsy and there were necklace fibers and abnormal triad structure localization on histopathology, consistent with myotubular myopathy. Whole genome sequencing revealed a premature stop codon in exon 13 of MTM1 (ChrX: 118,903,496 C > T, c.1467C>T, p.Arg512X). The puppy was humanely euthanized at 5 months of age. The puppy's dam was heterozygous for the variant, and 3 male puppies from a subsequent litter all of which died by 2 weeks of age were hemizygous for the variant. This naturally occurring mutation in Boykin spaniels causes a severe form of X-linked myotubular myopathy, comparable to the human counterpart.
Collapse
|
23
|
Schartner V, Laporte J, Böhm J. Abnormal Excitation-Contraction Coupling and Calcium Homeostasis in Myopathies and Cardiomyopathies. J Neuromuscul Dis 2020; 6:289-305. [PMID: 31356215 DOI: 10.3233/jnd-180314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Muscle contraction requires specialized membrane structures with precise geometry and relies on the concerted interplay of electrical stimulation and Ca2+ release, known as excitation-contraction coupling (ECC). The membrane structure hosting ECC is called triad in skeletal muscle and dyad in cardiac muscle, and structural or functional defects of triads and dyads have been observed in a variety of myopathies and cardiomyopathies. Based on their function, the proteins localized at the triad/dyad can be classified into three molecular pathways: the Ca2+ release complex (CRC), store-operated Ca2+ entry (SOCE), and membrane remodeling. All three are mechanistically linked, and consequently, aberrations in any of these pathways cause similar disease entities. This review provides an overview of the clinical and genetic spectrum of triad and dyad defects with a main focus of attention on the underlying pathomechanisms.
Collapse
Affiliation(s)
- Vanessa Schartner
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France.,INSERM U1258, Illkirch, France.,CNRS UMR7104, Illkirch, France.,Strasbourg University, Illkirch, France
| |
Collapse
|
24
|
Graham RJ, Ward E. X-linked myotubular myopathy and pulmonary blebs: Not just a muscle disorder. Muscle Nerve 2019; 60:E36-E38. [PMID: 31495930 DOI: 10.1002/mus.26697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Robert J Graham
- Department of Anesthesiology, Critical Care and Pain Medicine, Division of Critical Care, Boston Children's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Erin Ward
- MTM-CNM Family Connection, Inc, Methuen, Massachusetts
| |
Collapse
|
25
|
Kamio K, Takahashi Y, Ishihara K, Sekiya A, Kato S, Shimanuki I, Ide M, Furuoka H. Centronuclear Myopathy with Abundant Nemaline Rods in a Japanese Black and Hereford Crossbred Calf. J Comp Pathol 2019; 174:8-12. [PMID: 31955807 DOI: 10.1016/j.jcpa.2019.10.010] [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: 06/04/2019] [Revised: 09/17/2019] [Accepted: 10/19/2019] [Indexed: 10/25/2022]
Abstract
Histopathological examination was performed on skeletal and diaphragmatic muscles from an 8-month-old male crossbred calf showing abnormal gait and tremor of the hindlimbs. There were numerous round fibres with centrally placed nuclei forming nuclear chains in longitudinal sections, associated with interstitial fibrosis or adipose tissue infiltration. On nicotinamide adenine dinucleotide tetrazolium reductase (NADH-TR) staining, some muscle fibres in severe lesions showed a spoke-like appearance due to a radial arrangement of sarcoplasmic strands. Additionally, increased NADH-TR activity in the subsarcolemmal structures, appearingas ring-like or necklace-like forms, were observed. Transmission electron microscopy revealed dilated sarcoplasmic reticulum and variably shaped electron-dense inclusions consisting of myofibrillar streams. Another prominent feature was the existence of numerous nemaline rods within muscle fibres; these were stained red by Gomori's trichrome stain. Immunohistochemistry revealed that the nemaline rods showed strong immunoreactivity with α-actinin and desmin antibodies. Electron microscopically, these structures were composed of dense-homogeneous material and continuous with the Z disk. The case was diagnosed as centronuclear myopathy with increased nemaline rods.
Collapse
Affiliation(s)
- K Kamio
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan
| | - Y Takahashi
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan
| | - K Ishihara
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan
| | - A Sekiya
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan
| | - S Kato
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan
| | - I Shimanuki
- Tokachi Agricultural Mutual Aid Association, Obihiro, Japan
| | - M Ide
- Tokachi Agricultural Mutual Aid Association, Obihiro, Japan
| | - H Furuoka
- Division of Veterinary Sciences, Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Japan.
| |
Collapse
|
26
|
Granger N, Luján Feliu-Pascual A, Spicer C, Ricketts S, Hitti R, Forman O, Hersheson J, Houlden H. Charcot-Marie-Tooth type 4B2 demyelinating neuropathy in miniature Schnauzer dogs caused by a novel splicing SBF2 (MTMR13) genetic variant: a new spontaneous clinical model. PeerJ 2019; 7:e7983. [PMID: 31772832 PMCID: PMC6875392 DOI: 10.7717/peerj.7983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 10/02/2019] [Indexed: 01/23/2023] Open
Abstract
Background Charcot-Marie-Tooth (CMT) disease is the most common neuromuscular disorder in humans affecting 40 out of 100,000 individuals. In 2008, we described the clinical, electrophysiological and pathological findings of a demyelinating motor and sensory neuropathy in Miniature Schnauzer dogs, with a suspected autosomal recessive mode of inheritance based on pedigree analysis. The discovery of additional cases has followed this work and led to a genome-wide association mapping approach to search for the underlying genetic cause of the disease. Methods For genome wide association screening, genomic DNA samples from affected and unaffected dogs were genotyped using the Illumina CanineHD SNP genotyping array. SBF2 and its variant were sequenced using primers and PCRs. RNA was extracted from muscle of an unaffected and an affected dog and RT-PCR performed. Immunohistochemistry for myelin basic protein was performed on peripheral nerve section specimens. Results The genome-wide association study gave an indicative signal on canine chromosome 21. Although the signal was not of genome-wide significance due to the small number of cases, the SBF2 (also known as MTMR13) gene within the region of shared case homozygosity was a strong positional candidate, as 22 genetic variants in the gene have been associated with demyelinating forms of Charcot-Marie-Tooth disease in humans. Sequencing of SBF2 in cases revealed a splice donor site genetic variant, resulting in cryptic splicing and predicted early termination of the protein based on RNA sequencing results. Conclusions This study reports the first genetic variant in Miniature Schnauzer dogs responsible for the occurrence of a demyelinating peripheral neuropathy with abnormally folded myelin. This discovery establishes a genotype/phenotype correlation in affected Miniature Schnauzers that can be used for the diagnosis of these dogs. It further supports the dog as a natural model of a human disease; in this instance, Charcot-Marie-Tooth disease. It opens avenues to search the biological mechanisms responsible for the disease and to test new therapies in a non-rodent large animal model. In particular, recent gene editing methods that led to the restoration of dystrophin expression in a canine model of muscular dystrophy could be applied to other canine models such as this before translation to humans.
Collapse
Affiliation(s)
- Nicolas Granger
- Royal Veterinary College, University of London, Hatfield, United Kingdom.,Bristol Veterinary Specialists, CVS Referrals, Bristol, United Kingdom
| | | | - Charlotte Spicer
- Department of Molecular Neuroscience, UCL Institute of Neurology & National Hospital for Neurology and Neurosurgery & London, London, United Kingdom
| | - Sally Ricketts
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, United Kingdom
| | - Rebekkah Hitti
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, United Kingdom
| | - Oliver Forman
- Kennel Club Genetics Centre, Animal Health Trust, Newmarket, United Kingdom
| | - Joshua Hersheson
- Department of Molecular Neuroscience, UCL Institute of Neurology & National Hospital for Neurology and Neurosurgery & London, London, United Kingdom
| | - Henry Houlden
- Department of Molecular Neuroscience, UCL Institute of Neurology & National Hospital for Neurology and Neurosurgery & London, London, United Kingdom
| |
Collapse
|
27
|
Dupont JB, Guo J, Renaud-Gabardos E, Poulard K, Latournerie V, Lawlor MW, Grange RW, Gray JT, Buj-Bello A, Childers MK, Mack DL. AAV-Mediated Gene Transfer Restores a Normal Muscle Transcriptome in a Canine Model of X-Linked Myotubular Myopathy. Mol Ther 2019; 28:382-393. [PMID: 31784415 DOI: 10.1016/j.ymthe.2019.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 09/13/2019] [Accepted: 10/31/2019] [Indexed: 12/27/2022] Open
Abstract
Multiple clinical trials employing recombinant adeno-associated viral (rAAV) vectors have been initiated for neuromuscular disorders, including Duchenne and limb-girdle muscular dystrophies, spinal muscular atrophy, and recently X-linked myotubular myopathy (XLMTM). Our previous work on a canine model of XLMTM showed that a single rAAV8-cMTM1 systemic infusion corrected structural abnormalities within the muscle and restored contractile function, with affected dogs surviving more than 4 years post injection. This remarkable therapeutic efficacy presents a unique opportunity to identify the downstream molecular drivers of XLMTM pathology and to what extent the whole muscle transcriptome is restored to normal after gene transfer. Herein, RNA-sequencing was used to examine the transcriptomes of the Biceps femoris and Vastus lateralis in a previously described canine cohort that showed dose-dependent clinical improvements after rAAV8-cMTM1 gene transfer. Our analysis confirmed several dysregulated genes previously observed in XLMTM mice but also identified transcripts linked to XLMTM pathology. We demonstrated XLMTM transcriptome remodeling and dose-dependent normalization of gene expression after gene transfer and created metrics to pinpoint potential biomarkers of disease progression and correction.
Collapse
Affiliation(s)
- Jean-Baptiste Dupont
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - Jianjun Guo
- Audentes Therapeutics, San Francisco, CA 94108, USA
| | - Edith Renaud-Gabardos
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Karine Poulard
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Virginie Latournerie
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Michael W Lawlor
- Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - John T Gray
- Audentes Therapeutics, San Francisco, CA 94108, USA
| | - Ana Buj-Bello
- Genethon, INSERM UMR S951, Université Evry Val-d'Essone, Université Paris-Saclay, 91000 Evry, France
| | - Martin K Childers
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| | - David L Mack
- Department of Rehabilitation Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
28
|
Volpatti JR, Al-Maawali A, Smith L, Al-Hashim A, Brill JA, Dowling JJ. The expanding spectrum of neurological disorders of phosphoinositide metabolism. Dis Model Mech 2019; 12:12/8/dmm038174. [PMID: 31413155 PMCID: PMC6737944 DOI: 10.1242/dmm.038174] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Phosphoinositides (PIPs) are a ubiquitous group of seven low-abundance phospholipids that play a crucial role in defining localized membrane properties and that regulate myriad cellular processes, including cytoskeletal remodeling, cell signaling cascades, ion channel activity and membrane traffic. PIP homeostasis is tightly regulated by numerous inositol kinases and phosphatases, which phosphorylate and dephosphorylate distinct PIP species. The importance of these phospholipids, and of the enzymes that regulate them, is increasingly being recognized, with the identification of human neurological disorders that are caused by mutations in PIP-modulating enzymes. Genetic disorders of PIP metabolism include forms of epilepsy, neurodegenerative disease, brain malformation syndromes, peripheral neuropathy and congenital myopathy. In this Review, we provide an overview of PIP function and regulation, delineate the disorders associated with mutations in genes that modulate or utilize PIPs, and discuss what is understood about gene function and disease pathogenesis as established through animal models of these diseases. Summary: This Review highlights the intersection between phosphoinositides and the enzymes that regulate their metabolism, which together are crucial regulators of myriad cellular processes and neurological disorders.
Collapse
Affiliation(s)
- Jonathan R Volpatti
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Almundher Al-Maawali
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat 123, Oman
| | - Lindsay Smith
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Aqeela Al-Hashim
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada.,Department of Neuroscience, King Fahad Medical City, Riyadh 11525, Saudi Arabia
| | - Julie A Brill
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.,Program in Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - James J Dowling
- Division of Neurology and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada .,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
29
|
Abstract
The congenital myopathies are a genetically heterogeneous and diverse group of early-onset, nondystrophic neuromuscular disorders. While the originally reported "classical" entities within this group - Central Core Disease, Multiminicore Disease, Nemaline Myopathy, and Centronuclear Myopathy - were defined by the predominant finding on muscle biopsy, "novel" forms with multiple, subtle, and unusual histopathologic features have been described more recently, reflective of an expanding phenotypical spectrum. The main disease mechanisms concern excitation-contraction coupling, intracellular calcium homeostasis, and thin/thick filament interactions. Management to date has been mainly supportive. Therapeutic strategies currently at various stages of exploration include genetic interventions aimed at direct correction of the underlying genetic defect, enzyme replacement therapy, and pharmacologic approaches, either specifically targeting the principal effect of the underlying gene mutation, or addressing its downstream consequences more generally. Clinical trial development is accelerating but will require more robust natural history data and tailored outcome measures.
Collapse
Affiliation(s)
- Heinz Jungbluth
- Department of Paediatric Neurology, Neuromuscular Service, Evelina's Children Hospital, Guy's and St. Thomas' Hospital NHS Foundation Trust, London, United Kingdom; Randall Division for Cell and Molecular Biophysics, Muscle Signalling Section, London, United Kingdom; Department of Basic and Clinical Neuroscience, IoPPN, King's College, London, United Kingdom.
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital for Children, London, United Kingdom; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| |
Collapse
|
30
|
Tasfaout H, Cowling BS, Laporte J. Centronuclear myopathies under attack: A plethora of therapeutic targets. J Neuromuscul Dis 2019; 5:387-406. [PMID: 30103348 PMCID: PMC6218136 DOI: 10.3233/jnd-180309] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Centronuclear myopathies are a group of congenital myopathies characterized by severe muscle weakness, genetic heterogeneity, and defects in the structural organization of muscle fibers. Their names are derived from the central position of nuclei on biopsies, while they are at the fiber periphery under normal conditions. No specific therapy exists yet for these debilitating diseases. Mutations in the myotubularin phosphoinositides phosphatase, the GTPase dynamin 2, or amphiphysin 2 have been identified to cause respectively X-linked centronuclear myopathies (also called myotubular myopathy) or autosomal dominant and recessive forms. Mutations in additional genes, as RYR1, TTN, SPEG or CACNA1S, were linked to phenotypes that can overlap with centronuclear myopathies. Numerous animal models of centronuclear myopathies have been studied over the last 15 years, ranging from invertebrate to large mammalian models. Their characterization led to a partial understanding of the pathomechanisms of these diseases and allowed the recent validation of therapeutic proof-of-concepts. Here, we review the different therapeutic strategies that have been tested so far for centronuclear myopathies, some of which may be translated to patients.
Collapse
Affiliation(s)
- Hichem Tasfaout
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Belinda S. Cowling
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1258, Illkirch, France
- Centre National de la Recherche Scientifique (CNRS), UMR7104, Illkirch, France
- Université de Strasbourg, Illkirch, France
- Correspondence to: Jocelyn Laporte, Tel.: 33 0 388653412; E-mail:
| |
Collapse
|
31
|
Myotonia congenita in a Labrador Retriever with truncated CLCN1. Neuromuscul Disord 2018; 28:597-605. [DOI: 10.1016/j.nmd.2018.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/29/2018] [Accepted: 05/07/2018] [Indexed: 11/20/2022]
|
32
|
Danièle N, Moal C, Julien L, Marinello M, Jamet T, Martin S, Vignaud A, Lawlor MW, Buj-Bello A. Intravenous Administration of a MTMR2-Encoding AAV Vector Ameliorates the Phenotype of Myotubular Myopathy in Mice. J Neuropathol Exp Neurol 2018; 77:282-295. [PMID: 29408998 PMCID: PMC5939852 DOI: 10.1093/jnen/nly002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
X-linked myotubular myopathy (XLMTM) is a severe congenital disorder in male infants that leads to generalized skeletal muscle weakness and is frequently associated with fatal respiratory failure. XLMTM is caused by loss-of-function mutations in the MTM1 gene, which encodes myotubularin, the founder member of a family of 15 homologous proteins in mammals. We recently demonstrated the therapeutic efficacy of intravenous delivery of rAAV vectors expressing MTM1 in animal models of myotubular myopathy. Here, we tested whether the closest homologues of MTM1, MTMR1, and MTMR2 (the latter being implicated in Charcot-Marie-Tooth neuropathy type 4B1) are functionally redundant and could represent a therapeutic target for XLMTM. Serotype 9 recombinant AAV vectors encoding either MTM1, MTMR1, or MTMR2 were injected into the tibialis anterior muscle of Mtm1-deficient knockout mice. Two weeks after vector delivery, a therapeutic effect was observed with Mtm1 and Mtmr2, but not Mtmr1; with Mtm1 being the most efficacious transgene. Furthermore, intravenous administration of a single dose of the rAAV9-Mtmr2 vector in XLMTM mice improved the motor activity and muscle strength and prolonged survival throughout a 3-month study. These results indicate that strategies aiming at increasing MTMR2 expression levels in skeletal muscle may be beneficial in the treatment of myotubular myopathy.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Disease Models, Animal
- Escape Reaction/physiology
- HEK293 Cells
- Humans
- Locomotion/physiology
- Mice
- Muscle Contraction/drug effects
- Muscle Strength
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/ultrastructure
- Mutation
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/pathology
- Myopathies, Structural, Congenital/physiopathology
- Myopathies, Structural, Congenital/therapy
- PAX7 Transcription Factor/metabolism
- Phenotype
- Protein Tyrosine Phosphatases, Non-Receptor/administration & dosage
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Protein Tyrosine Phosphatases, Non-Receptor/metabolism
- RNA, Messenger/metabolism
- Transduction, Genetic
- Transfection
Collapse
Affiliation(s)
- Nathalie Danièle
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Christelle Moal
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Laura Julien
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Martina Marinello
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Thibaud Jamet
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Samia Martin
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Alban Vignaud
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| | - Michael W Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ana Buj-Bello
- INTEGRARE, INSERM UMRS 951, Univ Evry, Université Paris-Saclay, France
- R&D Department, Genethon, Evry, France
- Genethon, Evry, France
| |
Collapse
|
33
|
Pierson CR. Gene therapy strategies for X-linked myotubular myopathy. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1443807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Christopher R. Pierson
- Department of Pathology and Laboratory Medicine, Nationwide Children’s Hospital, Columbus, OH, USA
- Departments of Pathology and Biomedical Education & Anatomy, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
34
|
Single Intramuscular Injection of AAV-shRNA Reduces DNM2 and Prevents Myotubular Myopathy in Mice. Mol Ther 2018; 26:1082-1092. [PMID: 29506908 DOI: 10.1016/j.ymthe.2018.02.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/02/2018] [Accepted: 02/09/2018] [Indexed: 12/28/2022] Open
Abstract
Myotubular myopathy, or X-linked centronuclear myopathy, is a severe muscle disorder representing a significant burden for patients and their families. It is clinically characterized by neonatal and severe muscle weakness and atrophy. Mutations in the myotubularin (MTM1) gene cause myotubular myopathy, and no specific curative treatment is available. We previously found that dynamin 2 (DNM2) is upregulated in both Mtm1 knockout and patient muscle samples, whereas its reduction through antisense oligonucleotides rescues the clinical and histopathological features of this myopathy in mice. Here, we propose a novel approach targeting Dnm2 mRNA. We screened and validated in vitro and in vivo several short hairpin RNA (shRNA) sequences that efficiently target Dnm2 mRNA. A single intramuscular injection of AAV-shDnm2 resulted in long-term reduction of DNM2 protein level and restored muscle force, mass, histology, and myofiber ultrastructure and prevented molecular defects linked to the disease. Our results demonstrate a robust DNM2 knockdown and provide an alternative strategy based on reduction of DNM2 to treat myotubular myopathy.
Collapse
|
35
|
Felice KJ, Whitaker CH, Wu Q. Whole exome sequencing discloses a pathogenic MTM1 gene mutation and ends the diagnostic odyssey in an older woman with a progressive and seemingly sporadic myopathy: Case report and literature review of MTM1 manifesting female carriers. Neuromuscul Disord 2018; 28:339-345. [PMID: 29567349 DOI: 10.1016/j.nmd.2018.02.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 01/28/2023]
Abstract
We report the case of a 58-year-old woman with a progressive and seemingly sporadic myopathy who, later through whole exome sequencing, was diagnosed as a manifesting carrier of a myotubularin 1 gene mutation (c.342_342 + 4delAGTAA). As the case was a diagnostic challenge for 7 years, we thought it would be helpful to report the patient and review the other 25 cases thus far described. The manifesting carrier state is a rare cause for myopathic weakness in a female but should be strongly considered in kindreds with known affected males with myotubularin 1 gene mutations, and families with history of gestational polyhydramnios or male infantile death. Although the clinical phenotype is quite variable, the findings of ptosis, ophthalmoparesis, facial weakness, and asymmetrical limb involvement should raise the suspicion of the manifesting carrier state. Necklace fibers appear to be a highly sensitive and specific pathologic finding in such cases.
Collapse
Affiliation(s)
- Kevin J Felice
- Muscular Dystrophy Association Care Center, Department of Neuromuscular Medicine, Hospital for Special Care, New Britain, Connecticut, USA.
| | - Charles H Whitaker
- Muscular Dystrophy Association Care Center, Department of Neuromuscular Medicine, Hospital for Special Care, New Britain, Connecticut, USA
| | - Qian Wu
- Department of Pathology, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
36
|
Congenital myopathies: disorders of excitation-contraction coupling and muscle contraction. Nat Rev Neurol 2018; 14:151-167. [PMID: 29391587 DOI: 10.1038/nrneurol.2017.191] [Citation(s) in RCA: 191] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The congenital myopathies are a group of early-onset, non-dystrophic neuromuscular conditions with characteristic muscle biopsy findings, variable severity and a stable or slowly progressive course. Pronounced weakness in axial and proximal muscle groups is a common feature, and involvement of extraocular, cardiorespiratory and/or distal muscles can implicate specific genetic defects. Central core disease (CCD), multi-minicore disease (MmD), centronuclear myopathy (CNM) and nemaline myopathy were among the first congenital myopathies to be reported, and they still represent the main diagnostic categories. However, these entities seem to belong to a much wider phenotypic spectrum. To date, congenital myopathies have been attributed to mutations in over 20 genes, which encode proteins implicated in skeletal muscle Ca2+ homeostasis, excitation-contraction coupling, thin-thick filament assembly and interactions, and other mechanisms. RYR1 mutations are the most frequent genetic cause, and CCD and MmD are the most common subgroups. Next-generation sequencing has vastly improved mutation detection and has enabled the identification of novel genetic backgrounds. At present, management of congenital myopathies is largely supportive, although new therapeutic approaches are reaching the clinical trial stage.
Collapse
|
37
|
Beggs AH, Byrne BJ, De Chastonay S, Haselkorn T, Hughes I, James ES, Kuntz NL, Simon J, Swanson LC, Yang ML, Yu ZF, Yum SW, Prasad S. A multicenter, retrospective medical record review of X-linked myotubular myopathy: The recensus study. Muscle Nerve 2017; 57:550-560. [PMID: 29149770 PMCID: PMC5900738 DOI: 10.1002/mus.26018] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2017] [Indexed: 12/18/2022]
Abstract
Introduction: X‐linked myotubular myopathy (XLMTM), characterized by severe hypotonia, weakness, respiratory distress, and early mortality, is rare and natural history studies are few. Methods: RECENSUS is a multicenter chart review of male XLMTM patients characterizing disease burden and unmet medical needs. Data were collected between September 2014 and June 2016. Results: Analysis included 112 patients at six clinical sites. Most recent patient age recorded was ≤18 months for 40 patients and >18 months for 72 patients. Mean (SD) age at diagnosis was 3.7 (3.7) months and 54.3 (77.1) months, respectively. Mortality was 44% (64% ≤18 months; 32% >18 months). Premature delivery occurred in 34/110 (31%) births. Nearly all patients (90%) required respiratory support at birth. In the first year of life, patients underwent an average of 3.7 surgeries and spent 35% of the year in the hospital. Discussion: XLMTM is associated with high mortality, disease burden, and healthcare utilization. Muscle Nerve57: 550–560, 2018
Collapse
Affiliation(s)
- Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue Boston, Massachusetts, USA
| | - Barry J Byrne
- Children's Research Institute, University of Florida, Gainesville, Gainesville, Florida, USA
| | | | | | - Imelda Hughes
- Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Emma S James
- Audentes Therapeutics, San Francisco, California, USA
| | - Nancy L Kuntz
- Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
| | | | - Lindsay C Swanson
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue Boston, Massachusetts, USA
| | | | - Zi-Fan Yu
- Statistics Collaborative, Washington, DC
| | - Sabrina W Yum
- Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Suyash Prasad
- Audentes Therapeutics, San Francisco, California, USA
| |
Collapse
|
38
|
Elverman M, Goddard MA, Mack D, Snyder JM, Lawlor MW, Meng H, Beggs AH, Buj-Bello A, Poulard K, Marsh AP, Grange RW, Kelly VE, Childers MK. Long-term effects of systemic gene therapy in a canine model of myotubular myopathy. Muscle Nerve 2017; 56:943-953. [PMID: 28370029 DOI: 10.1002/mus.25658] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2017] [Indexed: 01/11/2023]
Abstract
INTRODUCTION X-linked myotubular myopathy (XLMTM), a devastating pediatric disease caused by the absence of the protein myotubularin, results from mutations in the MTM1 gene. While there is no cure for XLMTM, we previously reported effects of MTM1 gene therapy using adeno-associated virus (AAV) vector on muscle weakness and pathology in MTM1-mutant dogs. Here, we followed 2 AAV-infused dogs over 4 years. METHODS We evaluated gait, strength, respiration, neurological function, muscle pathology, AAV vector copy number (VCN), and transgene expression. RESULTS Four years following AAV-mediated gene therapy, gait, respiratory performance, neurological function and pathology in AAV-infused XLMTM dogs remained comparable to their healthy littermate controls despite a decline in VCN and muscle strength. CONCLUSIONS AAV-mediated gene transfer of MTM1 in young XLMTM dogs results in long-term expression of myotubularin transgene with normal muscular performance and neurological function in the absence of muscle pathology. These findings support a clinical trial in patients. Muscle Nerve 56: 943-953, 2017.
Collapse
Affiliation(s)
- Matthew Elverman
- Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Melissa A Goddard
- Department of Physiology and Pharmacology, School of Medicine, Wake Forest University Health Sciences, Winston-Salem, North Carolina, USA
| | - David Mack
- Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Campus Box 357340, Seattle, Washington, USA
| | - Michael W Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Hui Meng
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | - Anthony P Marsh
- Department of Health and Exercise Science, Wake Forest University, Winston-Salem, North Carolina, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Valerie E Kelly
- Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA
| | - Martin K Childers
- Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA.,Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
39
|
Mack DL, Poulard K, Goddard MA, Latournerie V, Snyder JM, Grange RW, Elverman MR, Denard J, Veron P, Buscara L, Le Bec C, Hogrel JY, Brezovec AG, Meng H, Yang L, Liu F, O'Callaghan M, Gopal N, Kelly VE, Smith BK, Strande JL, Mavilio F, Beggs AH, Mingozzi F, Lawlor MW, Buj-Bello A, Childers MK. Systemic AAV8-Mediated Gene Therapy Drives Whole-Body Correction of Myotubular Myopathy in Dogs. Mol Ther 2017; 25:839-854. [PMID: 28237839 DOI: 10.1016/j.ymthe.2017.02.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/13/2017] [Accepted: 02/01/2017] [Indexed: 12/18/2022] Open
Abstract
X-linked myotubular myopathy (XLMTM) results from MTM1 gene mutations and myotubularin deficiency. Most XLMTM patients develop severe muscle weakness leading to respiratory failure and death, typically within 2 years of age. Our objective was to evaluate the efficacy and safety of systemic gene therapy in the p.N155K canine model of XLMTM by performing a dose escalation study. A recombinant adeno-associated virus serotype 8 (rAAV8) vector expressing canine myotubularin (cMTM1) under the muscle-specific desmin promoter (rAAV8-cMTM1) was administered by simple peripheral venous infusion in XLMTM dogs at 10 weeks of age, when signs of the disease are already present. A comprehensive analysis of survival, limb strength, gait, respiratory function, neurological assessment, histology, vector biodistribution, transgene expression, and immune response was performed over a 9-month study period. Results indicate that systemic gene therapy was well tolerated, prolonged lifespan, and corrected the skeletal musculature throughout the body in a dose-dependent manner, defining an efficacious dose in this large-animal model of the disease. These results support the development of gene therapy clinical trials for XLMTM.
Collapse
MESH Headings
- Animals
- Biopsy
- Dependovirus/classification
- Dependovirus/genetics
- Disease Models, Animal
- Disease Progression
- Dogs
- Gait
- Gene Expression
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Genetic Vectors/administration & dosage
- Genetic Vectors/adverse effects
- Genetic Vectors/genetics
- Genetic Vectors/pharmacokinetics
- Immunity, Cellular
- Immunity, Humoral
- Kaplan-Meier Estimate
- Muscle Strength
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiopathology
- Muscle, Skeletal/ultrastructure
- Myopathies, Structural, Congenital/diagnosis
- Myopathies, Structural, Congenital/genetics
- Myopathies, Structural, Congenital/mortality
- Myopathies, Structural, Congenital/therapy
- Protein Tyrosine Phosphatases, Non-Receptor/genetics
- Recovery of Function
- Reflex
- Respiratory Function Tests
- Tissue Distribution
- Transgenes/genetics
- Transgenes/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA; Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98107, USA
| | - Karine Poulard
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France
| | - Melissa A Goddard
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98107, USA
| | | | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Robert W Grange
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Matthew R Elverman
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98107, USA
| | | | - Philippe Veron
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France
| | - Laurine Buscara
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France
| | | | - Jean-Yves Hogrel
- Neuromuscular Physiology and Evaluation Lab, Institut de Myologie, 75651 Paris, France
| | - Annie G Brezovec
- Department of Human Nutrition, Foods, and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Hui Meng
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Lin Yang
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Fujun Liu
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | | | - Nikhil Gopal
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98019, USA
| | - Valerie E Kelly
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA
| | - Barbara K Smith
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA
| | - Jennifer L Strande
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Fulvio Mavilio
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France
| | - Alan H Beggs
- The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Federico Mingozzi
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France; Institut de Myologie, University Pierre and Marie Curie, 75005 Paris, France
| | - Michael W Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ana Buj-Bello
- Genethon, 91000 Evry, France; INSERM, UMR_S951, 91002 Evry, France.
| | - Martin K Childers
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA; Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA 98107, USA.
| |
Collapse
|
40
|
Walmsley GL, Blot S, Venner K, Sewry C, Laporte J, Blondelle J, Barthélémy I, Maurer M, Blanchard-Gutton N, Pilot-Storck F, Tiret L, Piercy RJ. Progressive Structural Defects in Canine Centronuclear Myopathy Indicate a Role for HACD1 in Maintaining Skeletal Muscle Membrane Systems. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 187:441-456. [PMID: 27939133 DOI: 10.1016/j.ajpath.2016.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/12/2016] [Accepted: 10/17/2016] [Indexed: 11/16/2022]
Abstract
Mutations in HACD1/PTPLA cause recessive congenital myopathies in humans and dogs. Hydroxyacyl-coA dehydratases are required for elongation of very long chain fatty acids, and HACD1 has a role in early myogenesis, but the functions of this striated muscle-specific enzyme in more differentiated skeletal muscle remain unknown. Canine HACD1 deficiency is histopathologically classified as a centronuclear myopathy (CNM). We investigated the hypothesis that muscle from HACD1-deficient dogs has membrane abnormalities in common with CNMs with different genetic causes. We found progressive changes in tubuloreticular and sarcolemmal membranes and mislocalized triads and mitochondria in skeletal muscle from animals deficient in HACD1. Furthermore, comparable membranous abnormalities in cultured HACD1-deficient myotubes provide additional evidence that these defects are a primary consequence of altered HACD1 expression. Our novel findings, including T-tubule dilatation and disorganization, associated with defects in this additional CNM-associated gene provide a definitive pathophysiologic link with these disorders, confirm that dogs deficient in HACD1 are relevant models, and strengthen the evidence for a unifying pathogenesis in CNMs via defective membrane trafficking and excitation-contraction coupling in muscle. These results build on previous work by determining further functional roles of HACD1 in muscle and provide new insight into the pathology and pathogenetic mechanisms of HACD1 CNM. Consequently, alterations in membrane properties associated with HACD1 mutations should be investigated in humans with related phenotypes.
Collapse
Affiliation(s)
- Gemma L Walmsley
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom.
| | - Stéphane Blot
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Kerrie Venner
- Electron Microscopy Unit, Institute of Neurology, London, United Kingdom
| | - Caroline Sewry
- Dubowitz Neuromuscular Centre, University College London Institute of Child Health and Great Ormond Street Hospital, London, United Kingdom
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, Institute of Genetics and Cellular and Molecular Biology (IGBMC), Inserm U964, CNRS UMR7104, Strasbourg University, Illkirch, France
| | - Jordan Blondelle
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Inès Barthélémy
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Marie Maurer
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Nicolas Blanchard-Gutton
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Fanny Pilot-Storck
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Laurent Tiret
- French National Institute of Health and Medical Research (Inserm), Mondor Institute of Biomedical Research (IMRB) U955-E10 Biology of the Neuromuscular System, Créteil, France; University of Paris East, Alfort School of Veterinary Medicine (EnvA), Maisons-Alfort, France
| | - Richard J Piercy
- Comparative Neuromuscular Diseases Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, London, United Kingdom
| |
Collapse
|
41
|
Lawlor MW, Beggs AH, Buj-Bello A, Childers MK, Dowling JJ, James ES, Meng H, Moore SA, Prasad S, Schoser B, Sewry CA. Skeletal Muscle Pathology in X-Linked Myotubular Myopathy: Review With Cross-Species Comparisons. J Neuropathol Exp Neurol 2016; 75:102-10. [PMID: 26823526 PMCID: PMC4765322 DOI: 10.1093/jnen/nlv020] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
X-linked myotubular myopathy (XLMTM) is a devastating, rare, congenital myopathy caused by mutations in the MTM1 gene, resulting in a lack of or dysfunction of the enzyme myotubularin. This leads to severe perinatal weakness and distinctive muscle pathology. It was originally thought that XLMTM was related to developmental arrest in myotube maturation; however, the generation and characterization of several animal models have significantly improved our understanding of clinical and pathological aspects of this disorder. Myotubularin is now known to participate in numerous cellular processes including endosomal trafficking, excitation-contraction coupling, cytoskeletal organization, neuromuscular junction structure, autophagy, and satellite cell proliferation and survival. The available vertebrate models of XLMTM, which vary in severity from complete absence to reduced functional levels of myotubularin, recapitulate features of the human disease to a variable extent. Understanding how pathological endpoints in animals with XLMTM translate to human patients will be essential to interpret preclinical treatment trials and translate therapies into human clinical studies. This review summarizes the published animal models of XLMTM, including those of zebrafish, mice, and dogs, with a focus on their pathological features as compared to those seen in human XLMTM patients.
Collapse
|
42
|
Jungbluth H, Ochala J, Treves S, Gautel M. Current and future therapeutic approaches to the congenital myopathies. Semin Cell Dev Biol 2016; 64:191-200. [PMID: 27515125 DOI: 10.1016/j.semcdb.2016.08.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 12/14/2022]
Abstract
The congenital myopathies - including Central Core Disease (CCD), Multi-minicore Disease (MmD), Centronuclear Myopathy (CNM), Nemaline Myopathy (NM) and Congenital Fibre Type Disproportion (CFTD) - are a genetically heterogeneous group of early-onset neuromuscular conditions characterized by distinct histopathological features, and associated with a substantial individual and societal disease burden. Appropriate supportive management has substantially improved patient morbidity and mortality but there is currently no cure. Recent years have seen an exponential increase in the genetic and molecular understanding of these conditions, leading to the identification of underlying defects in proteins involved in calcium homeostasis and excitation-contraction coupling, thick/thin filament assembly and function, redox regulation, membrane trafficking and/or autophagic pathways. Based on these findings, specific therapies are currently being developed, or are already approaching the clinical trial stage. Despite undeniable progress, therapy development faces considerable challenges, considering the rarity and diversity of specific conditions, and the size and complexity of some of the genes and proteins involved. The present review will summarize the key genetic, histopathological and clinical features of specific congenital myopathies, and outline therapies already available or currently being developed in the context of known pathogenic mechanisms. The relevance of newly discovered molecular mechanisms and novel gene editing strategies for future therapy development will be discussed.
Collapse
Affiliation(s)
- Heinz Jungbluth
- Department of Paediatric Neurology, Neuromuscular Service, Evelina's Children Hospital, Guy's & St. Thomas' Hospital NHS Foundation Trust, London, United Kingdom; Randall Division for Cell and Molecular Biophysics, Muscle Signalling Section Biophysics and Cardiovascular Division, King's College BHF Centre of Research Excellence, United Kingdom; Department of Basic and Clinical Neuroscience, IoPPN, King's College, London, United Kingdom.
| | - Julien Ochala
- Centre of Human and Aerospace Physiological Sciences, King's College London, United Kingdom
| | - Susan Treves
- Departments of Biomedicine and Anaesthesia, Basel University Hospital, 4031 Basel, Switzerland
| | - Mathias Gautel
- Randall Division for Cell and Molecular Biophysics, Muscle Signalling Section Biophysics and Cardiovascular Division, King's College BHF Centre of Research Excellence, United Kingdom
| |
Collapse
|
43
|
Frase AR. Dream On: The Pursuit to Cure Myotubular Myopathy Born out of a Mother's Vision. Hum Gene Ther 2016; 27:341-4. [DOI: 10.1089/hum.2016.29024.arf] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
44
|
Polle F, Andrews FM, Gillon T, Eades SC, McConnico RS, Strain GM, Valberg SJ, Guo LT, Shelton GD. Suspected congenital centronuclear myopathy in an Arabian-cross foal. J Vet Intern Med 2016; 28:1886-91. [PMID: 25410957 PMCID: PMC4895639 DOI: 10.1111/jvim.12438] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 07/22/2014] [Accepted: 07/23/2014] [Indexed: 11/30/2022] Open
Affiliation(s)
- F Polle
- Equine Health Studies Program, Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mercier L, Böhm J, Fekonja N, Allio G, Lutz Y, Koch M, Goetz JG, Laporte J. In vivo imaging of skeletal muscle in mice highlights muscle defects in a model of myotubular myopathy. INTRAVITAL 2016; 5:e1168553. [PMID: 28243519 DOI: 10.1080/21659087.2016.1168553] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/09/2016] [Accepted: 03/10/2016] [Indexed: 12/11/2022]
Abstract
Skeletal muscle structure and function are altered in different myopathies. However, the understanding of the molecular and cellular mechanisms mainly rely on in vitro and ex vivo investigations in mammalian models. In order to monitor in vivo the intracellular structure of the neuromuscular system in its environment under normal and pathological conditions, we set-up and validated non-invasive imaging of ear and leg muscles in mice. This original approach allows simultaneous imaging of different cellular and intracellular structures such as neuromuscular junctions and sarcomeres, reconstruction of the 3D architecture of the neuromuscular system, and video recording of dynamic events such as spontaneous muscle fiber contraction. Second harmonic generation was combined with vital dyes and fluorescent-coupled molecules. Skin pigmentation, although limiting, did not prevent intravital imaging. Using this versatile toolbox on the Mtm1 knockout mouse, a model for myotubular myopathy which is a severe congenital myopathy in human, we identified several hallmarks of the disease such as defects in fiber size and neuromuscular junction shape. Intravital imaging of the neuromuscular system paves the way for the follow-up of disease progression or/and disease amelioration upon therapeutic tests. It has also the potential to reduce the number of animals needed to reach scientific conclusions.
Collapse
Affiliation(s)
- Luc Mercier
- Inserm U1109, MN3T, Strasbourg, France; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; LabEx Medalis, Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Johann Böhm
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Collège de France, Chaire de Génétique Humaine, Illkirch, France
| | - Nina Fekonja
- Inserm U1109, MN3T, Strasbourg, France; LabEx Medalis, Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Guillaume Allio
- Inserm U1109, MN3T, Strasbourg, France; LabEx Medalis, Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Yves Lutz
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
| | - Marc Koch
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France
| | - Jacky G Goetz
- Inserm U1109, MN3T, Strasbourg, France; LabEx Medalis, Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Collège de France, Chaire de Génétique Humaine, Illkirch, France
| |
Collapse
|
46
|
Snyder JM, Meisner A, Mack D, Goddard M, Coulter IT, Grange R, Childers MK. Validity of a Neurological Scoring System for Canine X-Linked Myotubular Myopathy. HUM GENE THER CL DEV 2016; 26:131-7. [PMID: 26086764 DOI: 10.1089/humc.2015.049] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
A simple clinical neurological test was developed to evaluate response to gene therapy in a preclinical canine model of X-linked myotubular myopathy (XLMTM). This devastating congenital myopathy is caused by mutation in the myotubularin (MTM1) gene. Clinical signs include muscle weakness, early respiratory failure, and ventilator dependence. A spontaneously occurring canine model has a similar clinical picture and histological abnormalities on muscle biopsy compared with patients. We developed a neuromuscular assessment score, graded on a scale from 10 (normal) to 1 (unable to maintain sternal recumbency). We hypothesize that this neurological assessment score correlates with genotype and established measures of disease severity and is reliable when performed by an independent observer. At 17 weeks of age, there was strong correlation between neurological assessment scores and established methods of severity testing. The neurological severity score correctly differentiated between XLMTM and wild-type dogs with good interobserver reliability, on the basis of strong agreement between neurological scores assigned by independent observers. Together, these data indicate that the neurological scoring system developed for this canine congenital neuromuscular disorder is reliable and valid. This scoring system may be helpful in evaluating response to therapy in preclinical testing in this disease model, such as response to gene therapy.
Collapse
Affiliation(s)
- Jessica M Snyder
- 1 Department of Comparative Medicine, University of Washington , Seattle, WA 98195
| | - Allison Meisner
- 2 Department of Biostatistics, University of Washington , Seattle, WA 98195
| | - David Mack
- 3 Department of Rehabilitation Medicine, University of Washington , Seattle, WA 98195.,4 Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington , Seattle, WA 98195
| | - Melissa Goddard
- 4 Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington , Seattle, WA 98195.,5 Department of Integrative Physiology and Pharmacology, Wake Forest University , Winston Salem, NC 27101
| | - Ian T Coulter
- 4 Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington , Seattle, WA 98195
| | - Robert Grange
- 6 Department of Human Nutrition, Foods and Exercise, Virginia Tech University , Blacksburg, VA 24061
| | - Martin K Childers
- 3 Department of Rehabilitation Medicine, University of Washington , Seattle, WA 98195.,4 Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington , Seattle, WA 98195
| |
Collapse
|
47
|
Cerda-Gonzalez S, Talarico L, Todhunter R. Noninvasive Assessment of Neuromuscular Disease in Dogs: Use of the 6-minute Walk Test to Assess Submaximal Exercise Tolerance in Dogs with Centronuclear Myopathy. J Vet Intern Med 2016; 30:808-12. [PMID: 27012153 PMCID: PMC4913584 DOI: 10.1111/jvim.13939] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 12/22/2015] [Accepted: 03/03/2016] [Indexed: 12/04/2022] Open
Abstract
Background Noninvasive methods of quantitating exercise tolerance in dogs with neuromuscular disease are needed both for clinical and research use. The 6‐minute walk test (6MWT) has been validated as a reliable test of exercise tolerance in dogs with pulmonary and cardiac disease, but not in dogs with neuromuscular disease. Hypothesis/Objectives Distance walked and number of steps taken during 6MWT will differ between Labrador retriever dogs with centronuclear myopathy (CNM) and control (ie, healthy) littermates. Animals Eight purebred Labrador retrievers were drawn from a purpose‐bred research colony (status: 3 clear, 2 carrier, and 3 homozygous mutants for the protein tyrosine phosphatase‐like A (PTPLA) gene mutation associated with CNM). Methods Pilot, prospective, Case–controlled study. Researchers were blinded to disease status. Each dog was leash‐trained and acclimatized to the testing area (length, 12.8 m). At the start of testing, each animal was fitted with a pedometer, a timer was started, and dogs were allowed to walk at their own pace for 6 minutes. Distance walked and pedometer readings were recorded. Results Degree of paresis varied among affected dogs, and was reflected by significant differences in distance walked between CNM‐affected dogs and those with clear and carrier genotypes (P = .048). Pedometer readings did not vary according to genotype (P = .86). Conclusions The 6MWT appears to differentiate between the ambulatory capacity of normal and CNM‐affected dogs. Additional studies are needed to confirm this relationship in a larger number of dogs, and to evaluate the ability of the 6MWT to differentiate between dogs with variable severity of neuromuscular disease‐associated exercise intolerance.
Collapse
Affiliation(s)
- S Cerda-Gonzalez
- Department of Clinical Sciences (College of Veterinary Medicine), Cornell University, Ithaca, NY
| | - L Talarico
- SouthPaws Veterinary Specialists and Emergency Center, Fairfax, VA
| | - R Todhunter
- Department of Clinical Sciences (College of Veterinary Medicine), Cornell University, Ithaca, NY
| |
Collapse
|
48
|
Canine-Inherited Dystrophinopathies and Centronuclear Myopathies. REGENERATIVE MEDICINE FOR DEGENERATIVE MUSCLE DISEASES 2016. [DOI: 10.1007/978-1-4939-3228-3_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
49
|
Goddard MA, Mack DL, Czerniecki SM, Kelly VE, Snyder JM, Grange RW, Lawlor MW, Smith BK, Beggs AH, Childers MK. Muscle pathology, limb strength, walking gait, respiratory function and neurological impairment establish disease progression in the p.N155K canine model of X-linked myotubular myopathy. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:262. [PMID: 26605308 DOI: 10.3978/j.issn.2305-5839.2015.10.31] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Loss-of-function mutations in the myotubularin (MTM1) gene cause X-linked myotubular myopathy (XLMTM), a fatal, inherited pediatric disease that affects the entire skeletal musculature. Labrador retriever dogs carrying an MTM1 missense mutation exhibit strongly reduced synthesis of myotubularin, the founder member of a lipid phosphatase required for normal skeletal muscle function. The resulting canine phenotype resembles that of human patients with comparably severe mutations, and survival does not normally exceed 4 months. METHODS We studied MTM1 mutant dogs (n=7) and their age-matched control littermates (n=6) between the ages of 10 and 25 weeks. Investigators blinded to the animal identities sequentially measured limb muscle pathology, fore- and hind limb strength, walking gait, respiratory function and neurological impairment. RESULTS MTM1-mutant puppies display centrally-nucleated myofibers of reduced size and disrupted sarcotubular architecture progressing until the end of life, an average of 17 weeks. In-life measures of fore- and hind limb strength establish the rate at which XLMTM muscles weaken, and their corresponding decrease in gait velocity and stride length. Pulmonary function tests in affected dogs reveal a right-shifted relationship between peak inspiratory flow (PIF) and inspiratory time (TI); neurological assessments indicate that affected puppies as young as 10 weeks show early signs of neurological impairment (neurological severity score, NSS =8.6±0.9) with progressive decline (NSS =5.6±1.7 at 17 weeks-of-age). CONCLUSIONS Our findings document the rate of disease progression in a large animal model of XLMTM and lay a foundation for preclinical studies.
Collapse
Affiliation(s)
- Melissa A Goddard
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - David L Mack
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan M Czerniecki
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Valerie E Kelly
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jessica M Snyder
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert W Grange
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael W Lawlor
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Barbara K Smith
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan H Beggs
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Martin K Childers
- 1 Institute for Stem Cell and Regenerative Medicine, 2 Department of Rehabilitation Medicine, School of Medicine, University of Washington, Seattle, Washington, USA ; 3 Department of Comparative Medicine, University of Washington, Seattle, Washington, USA ; 4 Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic and State University, Blacksburg, Virginia, USA ; 5 Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI, USA ; 6 Department of Physical Therapy, University of Florida, Gainesville, FL, USA ; 7 The Manton Center for Orphan Disease Research, Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
50
|
Mhlanga-Mutangadura T, Johnson GS, Schnabel RD, Taylor JF, Johnson GC, Katz ML, Shelton GD, Lever TE, Giuliano E, Granger N, Shomper J, O'Brien DP. A mutation in the Warburg syndrome gene, RAB3GAP1, causes a similar syndrome with polyneuropathy and neuronal vacuolation in Black Russian Terrier dogs. Neurobiol Dis 2015; 86:75-85. [PMID: 26607784 DOI: 10.1016/j.nbd.2015.11.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 11/30/2022] Open
Abstract
An autosomal recessive disease of Black Russian Terriers was previously described as a juvenile-onset, laryngeal paralysis and polyneuropathy similar to Charcot Marie Tooth disease in humans. We found that in addition to an axonal neuropathy, affected dogs exhibit microphthalmia, cataracts, and miotic pupils. On histopathology, affected dogs exhibit a spongiform encephalopathy characterized by accumulations of abnormal, membrane-bound vacuoles of various sizes in neuronal cell bodies, axons and adrenal cells. DNA from an individual dog with this polyneuropathy with ocular abnormalities and neuronal vacuolation (POANV) was used to generate a whole genome sequence which contained a homozygous RAB3GAP1:c.743delC mutation that was absent from 73 control canine whole genome sequences. An additional 12 Black Russian Terriers with POANV were RAB3GAP1:c.743delC homozygotes. DNA samples from 249 Black Russian Terriers with no known signs of POANV were either heterozygotes or homozygous for the reference allele. Mutations in human RAB3GAP1 cause Warburg micro syndrome (WARBM), a severe developmental disorder characterized by abnormalities of the eye, genitals and nervous system including a predominantly axonal peripheral neuropathy. RAB3GAP1 encodes the catalytic subunit of a GTPase activator protein and guanine exchange factor for Rab3 and Rab18 respectively. Rab proteins are involved in membrane trafficking in the endoplasmic reticulum, axonal transport, autophagy and synaptic transmission. The neuronal vacuolation and membranous inclusions and vacuoles in axons seen in this canine disorder likely reflect alterations of these processes. Thus, this canine disease could serve as a model for WARBM and provide insight into its pathogenesis and treatment.
Collapse
Affiliation(s)
| | - Gary S Johnson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, USA
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, USA; Informatics Institute, University of Missouri, Columbia, USA
| | - Jeremy F Taylor
- Division of Animal Sciences, University of Missouri, Columbia, USA
| | - Gayle C Johnson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, USA
| | - Martin L Katz
- Mason Eye Institute, University of Missouri, Columbia, USA
| | - G Diane Shelton
- Department of Pathology, University of California, San Diego, La Jolla, USA
| | - Teresa E Lever
- Department of Otolaryngology, University of Missouri, Columbia, USA
| | - Elizabeth Giuliano
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA
| | - Nicolas Granger
- School of Veterinary Sciences, University of Bristol, Langford House, Langford, Somerset BS40 5DU, UK
| | - Jeremy Shomper
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA
| | - Dennis P O'Brien
- Department of Veterinary Medicine & Surgery, University of Missouri, Columbia, USA.
| |
Collapse
|