1
|
Gandhi S, Sweeney HL, Hart CC, Han R, Perry CGR. Cardiomyopathy in Duchenne Muscular Dystrophy and the Potential for Mitochondrial Therapeutics to Improve Treatment Response. Cells 2024; 13:1168. [PMID: 39056750 PMCID: PMC11274633 DOI: 10.3390/cells13141168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Cora C. Hart
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
2
|
Mantle D, Hargreaves IP. Efficacy and Safety of Coenzyme Q10 Supplementation in Neonates, Infants and Children: An Overview. Antioxidants (Basel) 2024; 13:530. [PMID: 38790635 PMCID: PMC11117623 DOI: 10.3390/antiox13050530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
To date, there have been no review articles specifically relating to the general efficacy and safety of coenzyme Q10 (CoQ10) supplementation in younger subjects. In this article, we therefore reviewed the efficacy and safety of CoQ10 supplementation in neonates (less than 1 month of age), infants (up to 1 year of age) and children (up to 12 years of age). As there is no rationale for the supplementation of CoQ10 in normal younger subjects (as there is in otherwise healthy older subjects), all of the articles in the medical literature reviewed in the present article therefore refer to the supplementation of CoQ10 in younger subjects with a variety of clinical disorders; these include primary CoQ10 deficiency, acyl CoA dehydrogenase deficiency, Duchenne muscular dystrophy, migraine, Down syndrome, ADHD, idiopathic cardiomyopathy and Friedreich's ataxia.
Collapse
Affiliation(s)
- David Mantle
- Pharma Nord (UK) Ltd., Morpeth, Northumberland NE61 2DB, UK
| | - Iain Parry Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Merseyside L3 5UX, UK;
| |
Collapse
|
3
|
Naarding KJ, Stimpson G, Ward SJ, Goemans N, McDonald C, Mercuri E, Muntoni F. 269th ENMC international workshop: 10 years of clinical trials in Duchenne muscular dystrophy - What have we learned? 9-11 December 2022, Hoofddorp, The Netherlands. Neuromuscul Disord 2023; 33:897-910. [PMID: 37926638 DOI: 10.1016/j.nmd.2023.10.003] [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: 09/30/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
There are multiple avenues for therapeutic development in Duchenne muscular dystrophy (DMD), which are highlighted in the first section of this report for the "10 years of Clinical trials in DMD - What have we learned?" workshop. This report then provides an overview of the presentations made at the workshop grouped into the following core themes: trial outcomes, disease heterogeneity, meaningfulness of outcomes and the utility of real-world data in trials. Finally, we present the consensus that was achieved at the workshop on the learning points from 10 years of clinical trials in DMD, and possible action points from these. This includes further work in expanding the scope and range of trial outcomes and assessing the efficacy of new trial structures for DMD. We also highlight several points which should be addressed during future interactions with regulators, such as clinical meaningfulness and the use of real-world data.
Collapse
Affiliation(s)
- Karin J Naarding
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands; Duchenne Center Netherlands, the Netherlands
| | - Georgia Stimpson
- UCL Great Ormond Street Institute of Child Health, Dubowitz Neuromuscular Centre, London, UK
| | - Susan J Ward
- Collaborative Trajectory Analysis Project (cTAP), United States
| | - Nathalie Goemans
- University Hospitals Leuven, Dept of Child Neurology, Leuven, Belgium
| | - Craig McDonald
- Department of Physical Medicine and Rehabilitation in Sacramento, University of California, Davis, CA, United States
| | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University, Rome, Italy; Centro Clinico Nemo, U.O.C. Neuropsichiatria Infantile Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, Dubowitz Neuromuscular Centre, London, UK; National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
4
|
Yu W, Jiang W, Wu W, Wang G, Zhao D, Yan C, Lin P. Combining idebenone and rosuvastatin prevents atherosclerosis by suppressing oxidative stress and NLRP3 inflammasome activation. Eur J Pharmacol 2023; 955:175911. [PMID: 37451421 DOI: 10.1016/j.ejphar.2023.175911] [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: 02/12/2023] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Atherosclerosis is a progressive inflammatory disease activated by excessive oxidized low-density lipoprotein (ox-LDL). Statins are the first-line choice to reduce the risk of cardiovascular disease. However, statin-associated side effects prompt dose reduction or discontinuation. Idebenone could protect against atherosclerosis by scavenging reactive oxygen species (ROS). Although both idebenone and statins have certain efficacy, neither of them can achieve a completely satisfactory effect. Here, we aim to investigate the anti-atherosclerotic effect of the combination of idebenone and statins. Apolipoprotein E knockout (ApoE-/-) mice were given idebenone (400 mg/kg/d), rosuvastatin (10 mg/kg/d) or a combination of idebenone and rosuvastatin. Histological and immunohistochemical staining were used to analyze the size and composition of the plaque. In vivo and in vitro experiments were conducted to explore the possible mechanism. Idebenone and rosuvastatin both reduced plaque burden and increased the stability of atherosclerotic plaques in the ApoE-/- mice. Mice receiving the combination therapy had even reduced and more stable atherosclerotic plaques than mice treated with idebenone or rosuvastatin alone. NLRP3 and IL-1β were further downregulated in mice receiving combination therapy compared with mice treated with monotherapy. The combination treatment also markedly reduced oxidative stress and cell apoptosis in vivo and in vitro. In conclusion, our data demonstrate that the combination of idebenone and rosuvastatin works synergistically to inhibit atherosclerosis, and that the use of both substances together is more effective than using either substance alone. From a therapeutic point, combining idebenone and rosuvastatin appears to be a promising strategy to further prevent atherosclerosis.
Collapse
Affiliation(s)
- Wenfei Yu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China; University of Health and Rehabilitation Sciences, No. 17, Shandong Road, Shinan District, Qingdao City, Shandong Province, China
| | - Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong Province, China
| | - Wenjing Wu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Guangyu Wang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Dandan Zhao
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China
| | - Chuanzhu Yan
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| | - Pengfei Lin
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
5
|
Juracic ES, Brahmbhatt AN, Hawke TJ. Therapeutic targeting of mitochondria in muscular dystrophies. Editorial focus on: "Mitochondrial creatine sensitivity is lost in the D2. mdx model of Duchenne muscular dystrophy and rescued by the mitochondrial-enhancing compound Olesoxime". Am J Physiol Cell Physiol 2023; 325:C563-C564. [PMID: 37458438 DOI: 10.1152/ajpcell.00301.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 08/09/2023]
Affiliation(s)
- Emma S Juracic
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Aditya N Brahmbhatt
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
6
|
Li D, Zhang G, Wang Z, Guo J, Liu Y, Lu Y, Qin Z, Xu Y, Cao C, Wang B, Guo Q, Wang Y, Liu G, Cui X, Zhang J, Tang J. Idebenone attenuates ferroptosis by inhibiting excessive autophagy via the ROS-AMPK-mTOR pathway to preserve cardiac function after myocardial infarction. Eur J Pharmacol 2023; 943:175569. [PMID: 36740037 DOI: 10.1016/j.ejphar.2023.175569] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of mortality worldwide. As a type of CVDs, myocardial infarction (MI) induces ischemia hypoxia, which leads to excessive reactive oxygen species (ROS), resulting in multiple cell deaths and contributing to the subsequent development of heart failure or premature death. Recent evidence indicates that ROS-induced lipid peroxidation promotes autophagy and ferroptosis, leading to the loss of healthy myocardium and resulting in the dysfunction of cardiac tissue. Theoretically, cardiac function would be preserved after MI by inhibiting autophagy and ferroptosis. As an analog of coenzyme Q10 (CoQ10) and a clinically approved drug, idebenone would be used to inhibit ferroptosis and preserve cardiac function due to its capacity to improve mitochondrial physiology with antioxidant and anti-inflammatory properties. Here, we confirmed that the addition of idebenone inhibited H2O2-induced and RSL3-induced ferroptosis. Furthermore, the ROS-AMPK-mTOR pathway axis was identified as the signaling pathway that idebenone stimulated to prevent excessive autophagy and consequent ferroptosis. In the MI animal model, idebenone demonstrated a cardioprotective role by regulating ROS-dependent autophagy and inhibiting ferroptosis, which paves the way for the future clinical translation of idebenone in MI management.
Collapse
Affiliation(s)
- Demin Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zeyu Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Jiacheng Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yu Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Yongzheng Lu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Zhen Qin
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yanyan Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Chang Cao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Bo Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Qianqian Guo
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Yunzhe Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China
| | - Guozhen Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518172, China.
| | - Jinying Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China.
| | - Junnan Tang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, 450018, China.
| |
Collapse
|
7
|
Valduga AH, Mizobuti DS, Moraes FDSR, Mâncio RD, Moraes LHR, Hermes TDA, Macedo AB, Minatel E. Protection of dystrophic muscle cells using Idebenone correlates with the interplay between calcium, oxidative stress and inflammation. Int J Exp Pathol 2023; 104:4-12. [PMID: 36565155 PMCID: PMC9845605 DOI: 10.1111/iep.12463] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/25/2022] Open
Abstract
There is strong cross-talk between abnormal intracellular calcium concentration, high levels of reactive oxygen species (ROS) and an exacerbated inflammatory process in the dystrophic muscles of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD). In this study, we investigated effects of Idebenone, a potent anti-oxidant, on oxidative stress markers, the anti-oxidant defence system, intracellular calcium concentrations and the inflammatory process in primary dystrophic muscle cells from mdx mice. Dystrophic muscle cells were treated with Idebenone (0.05 μM) for 24 h. The untreated mdx muscle cells were used as controls. The MTT assay showed that Idebenone did not have a cytotoxic effect on the dystrophic muscle cells. The Idebenone treatment was able to reduce the levels of oxidative stress markers, such as H2 O2 and 4-HNE, as well as decreasing intracellular calcium influx in the dystrophic muscle cells. Regarding Idebenone effects on the anti-oxidant defence system, an up-regulation of catalase levels, glutathione reductase (GR), glutathione peroxidase (GPx) and superoxide dismutase (SOD) activity was observed in the dystrophic muscle cells. In addition, the Idebenone treatment was also associated with reduction in inflammatory molecules, such as nuclear factor kappa-B (NF-κB) and tumour necrosis factor (TNF) in mdx muscle cells. These outcomes supported the use of Idebenone as a protective agent against oxidative stress and related signalling mechanisms involved in dystrophinopathies, such as DMD.
Collapse
Affiliation(s)
- Amanda Harduim Valduga
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Daniela Sayuri Mizobuti
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Fernanda dos Santos Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Rafael Dias Mâncio
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Luis Henrique Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Túlio de Almeida Hermes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Aline Barbosa Macedo
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| |
Collapse
|
8
|
Su Y, Song Y. The new challenge of “exercise + X″ therapy for Duchenne muscular dystrophy—Individualized identification of exercise tolerance and precise implementation of exercise intervention. Front Physiol 2022; 13:947749. [PMID: 35991169 PMCID: PMC9389311 DOI: 10.3389/fphys.2022.947749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive fatal muscular disease. Gene therapy, cell therapy, and drug therapy are currently the most widely used treatments for DMD. However, many experiments on animals and humans suggested that appropriate exercise could improve the effectiveness of such precision medicine treatment, thereby improving patient’s muscle quality and function. Due to the striated muscle damage of DMD individuals, there are still many debates about whether DMD animals or patients can exercise, how to exercise, when to exercise best, and how to exercise effectively. The purpose of this review is to summarize and investigate the scientific basis and efficacy of exercise as an adjuvant therapy for DMD gene therapy, cell therapy and drug therapy, as well as to present the theoretical framework and optional strategies of “exercise + X″″ combination therapy.
Collapse
Affiliation(s)
- Yuhui Su
- Department of Exercise Physiology, Beijing Sport University, Beijing, China
- Institute of Physical Education, Jilin Normal University, Siping, China
| | - Yafeng Song
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
- *Correspondence: Yafeng Song,
| |
Collapse
|
9
|
Rocha CT, Escolar DM. Treatment and Management of Muscular Dystrophies. Neuromuscul Disord 2022. [DOI: 10.1016/b978-0-323-71317-7.00020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
10
|
Markati T, De Waele L, Schara-Schmidt U, Servais L. Lessons Learned from Discontinued Clinical Developments in Duchenne Muscular Dystrophy. Front Pharmacol 2021; 12:735912. [PMID: 34790118 PMCID: PMC8591262 DOI: 10.3389/fphar.2021.735912] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 10/12/2021] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked condition caused by a deficiency of functional dystrophin protein. Patients experience progressive muscle weakness, cardiomyopathy and have a decreased life expectancy. Standards of care, including treatment with steroids, and multidisciplinary approaches have extended the life expectancy and improved the quality of life of patients. In the last 30 years, several compounds have been assessed in preclinical and clinical studies for their ability to restore functional dystrophin levels or to modify pathways involved in DMD pathophysiology. However, there is still an unmet need with regards to a disease-modifying treatment for DMD and the attrition rate between early-phase and late-phase clinical development remains high. Currently, there are 40 compounds in clinical development for DMD, including gene therapy and antisense oligonucleotides for exon skipping. Only five of them have received conditional approval in one jurisdiction subject to further proof of efficacy. In this review, we present data of another 16 compounds that failed to complete clinical development, despite positive results in early phases of development in some cases. We examine the reasons for the high attrition rate and we suggest solutions to avoid similar mistakes in the future.
Collapse
Affiliation(s)
- Theodora Markati
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Liesbeth De Waele
- KU Leuven Department of Development and Regeneration, Leuven, Belgium
- Department of Paediatric Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Urlike Schara-Schmidt
- Department of Pediatric Neurology, Center for Neuromuscular Diseases, Center for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, Essen, Germany
| | - Laurent Servais
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
- Division of Child Neurology, Reference Center for Neuromuscular Disease, Centre Hospitalier Régional de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège, Liège, Belgium
| |
Collapse
|
11
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
12
|
Yao S, Chen Z, Yu Y, Zhang N, Jiang H, Zhang G, Zhang Z, Zhang B. Current Pharmacological Strategies for Duchenne Muscular Dystrophy. Front Cell Dev Biol 2021; 9:689533. [PMID: 34490244 PMCID: PMC8417245 DOI: 10.3389/fcell.2021.689533] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked neuromuscular disorder caused by the absence of dystrophin protein, which is essential for muscle fiber integrity. Loss of dystrophin protein leads to recurrent myofiber damage, chronic inflammation, progressive fibrosis, and dysfunction of muscle stem cells. There is still no cure for DMD so far and the standard of care is principally limited to symptom relief through glucocorticoids treatments. Current therapeutic strategies could be divided into two lines. Dystrophin-targeted therapeutic strategies that aim at restoring the expression and/or function of dystrophin, including gene-based, cell-based and protein replacement therapies. The other line of therapeutic strategies aims to improve muscle function and quality by targeting the downstream pathological changes, including inflammation, fibrosis, and muscle atrophy. This review introduces the important developments in these two lines of strategies, especially those that have entered the clinical phase and/or have great potential for clinical translation. The rationale and efficacy of each agent in pre-clinical or clinical studies are presented. Furthermore, a meta-analysis of gene profiling in DMD patients has been performed to understand the molecular mechanisms of DMD.
Collapse
Affiliation(s)
- Shanshan Yao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
13
|
Rovira Gonzalez YI, Moyer AL, LeTexier NJ, Bratti AD, Feng S, Peña V, Sun C, Pulcastro H, Liu T, Iyer SR, Lovering RM, O'Rourke B, Wagner KR. Mss51 deletion increases endurance and ameliorates histopathology in the mdx mouse model of Duchenne muscular dystrophy. FASEB J 2021; 35:e21276. [PMID: 33423297 DOI: 10.1096/fj.202002106rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/25/2020] [Accepted: 11/30/2020] [Indexed: 11/11/2022]
Abstract
Mitochondrial derangement is an important contributor to the pathophysiology of muscular dystrophies and may be among the earliest cellular deficits. We have previously shown that disruption of Mss51, a mammalian skeletal muscle protein that localizes to the mitochondria, results in enhanced muscle oxygen consumption rate, increased endurance capacity, and improved limb muscle strength in mice with wildtype background. Here, we investigate whether Mss51 deletion in the mdx murine model of Duchenne muscular dystrophy (mdx-Mss51 KO) counteracts the muscle pathology and mitochondrial irregularities observed in mdx mice. We found that mdx-Mss51 KO mice had increased myofiber oxygen consumption rates and an amelioration of muscle histopathology compared to mdx counterparts. This corresponded with greater treadmill endurance and less percent fatigue in muscle physiology, but no improvement in forelimb grip strength or limb muscle force production. These findings suggest that although Mss51 deletion ameliorates the skeletal muscle mitochondrial respiration defects in mdx and improves fatigue resistance in vivo, the lack of improvement in force production suggests that this target alone may be insufficient for a therapeutic effect.
Collapse
Affiliation(s)
- Yazmin I Rovira Gonzalez
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Adam L Moyer
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Nicolas J LeTexier
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - August D Bratti
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Siyuan Feng
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Vanessa Peña
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Congshan Sun
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hannah Pulcastro
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Ting Liu
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shama R Iyer
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA.,Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
14
|
Uchimura T, Asano T, Nakata T, Hotta A, Sakurai H. A muscle fatigue-like contractile decline was recapitulated using skeletal myotubes from Duchenne muscular dystrophy patient-derived iPSCs. CELL REPORTS MEDICINE 2021; 2:100298. [PMID: 34195678 PMCID: PMC8233665 DOI: 10.1016/j.xcrm.2021.100298] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/28/2021] [Accepted: 05/10/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a muscle degenerating disease caused by dystrophin deficiency, for which therapeutic options are limited. To facilitate drug development, it is desirable to develop in vitro disease models that enable the evaluation of DMD declines in contractile performance. Here, we show MYOD1-induced differentiation of hiPSCs into functional skeletal myotubes in vitro with collagen gel and electrical field stimulation (EFS). Long-term EFS training (0.5 Hz, 20 V, 2 ms, continuous for 2 weeks) mimicking muscle overuse recapitulates declines in contractile performance in dystrophic myotubes. A screening of clinically relevant drugs using this model detects three compounds that ameliorate this decline. Furthermore, we validate the feasibility of adapting the model to a 96-well culture system using optogenetic technology for large-scale screening. Our results support a disease model using patient-derived iPSCs that allows for the recapitulation of the contractile pathogenesis of DMD and a screening strategy for drug development.
Collapse
Affiliation(s)
- Tomoya Uchimura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Toshifumi Asano
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Takao Nakata
- Department of Cell Biology, Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan.,The Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Akitsu Hotta
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| | - Hidetoshi Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.,Takeda-CiRA Joint Program, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
15
|
Spurney CF, Ascheim D, Charnas L, Cripe L, Hor K, King N, Kinnett K, McNally EM, Sauer JM, Sweeney L, Villa C, Markham LW. Current state of cardiac troponin testing in Duchenne muscular dystrophy cardiomyopathy: review and recommendations from the Parent Project Muscular Dystrophy expert panel. Open Heart 2021; 8:openhrt-2021-001592. [PMID: 33762424 PMCID: PMC7993361 DOI: 10.1136/openhrt-2021-001592] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/17/2021] [Accepted: 03/05/2021] [Indexed: 01/06/2023] Open
Abstract
Cardiac disease is now the leading cause of death in Duchenne muscular dystrophy (DMD). Clinical evaluations over time have demonstrated asymptomatic cardiac troponin elevations and acute elevations are associated with symptoms and cardiac dysfunction in DMD. Clinicians require a better understanding of the relationship of symptoms, troponin levels and progression of cardiac disease in DMD. As clinical trials begin to assess novel cardiac therapeutics in DMD, troponin levels in DMD are important for safety monitoring and outcome measures. The Parent Project Muscular Dystrophy convened an expert panel of cardiologists, scientists, and regulatory and industry specialists on 16 December 2019 in Silver Spring, Maryland and reviewed published and unpublished data from their institutions. The panel recommended retrospective troponin data analyses, prospective longitudinal troponin collection using high-sensitivity cardiac troponin I assays, inclusion of troponin in future clinical trial outcomes and future development of clinical guidelines for monitoring and treating troponin elevations in DMD.
Collapse
Affiliation(s)
- Christopher F Spurney
- Children's National Heart Institute, Children's National Hospital, Washington, District of Columbia, USA
| | | | - Lawrence Charnas
- Clinical Research Rare Neurology Disease, Pfizer, Cambridge, Massachusetts, USA
| | - Linda Cripe
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kan Hor
- The Heart Center, Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | - Kathi Kinnett
- Parent Project Muscular Dystrophy, Hackensack, New Jersey, USA
| | | | | | - Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Chet Villa
- Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Larry W Markham
- Pediatrics, Division of Pediatric Cardiology, Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| |
Collapse
|
16
|
Nitahara-Kasahara Y, Kuraoka M, Guillermo PH, Hayashita-Kinoh H, Maruoka Y, Nakamura-Takahasi A, Kimura K, Takeda S, Okada T. Dental pulp stem cells can improve muscle dysfunction in animal models of Duchenne muscular dystrophy. Stem Cell Res Ther 2021; 12:78. [PMID: 33494794 PMCID: PMC7831244 DOI: 10.1186/s13287-020-02099-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an inherited progressive disorder that causes skeletal and cardiac muscle deterioration with chronic inflammation. Dental pulp stem cells (DPSCs) are attractive candidates for cell-based strategies for DMD because of their immunosuppressive properties. Therefore, we hypothesized that systemic treatment with DPSCs might show therapeutic benefits as an anti-inflammatory therapy. Methods To investigate the potential benefits of DPSC transplantation for DMD, we examined disease progression in a DMD animal model, mdx mice, by comparing them with different systemic treatment conditions. The DPSC-treated model, a canine X-linked muscular dystrophy model in Japan (CXMDJ), which has a severe phenotype similar to that of DMD patients, also underwent comprehensive analysis, including histopathological findings, muscle function, and locomotor activity. Results We demonstrated a therapeutic strategy for long-term functional recovery in DMD using repeated DPSC administration. DPSC-treated mdx mice and CXMDJ showed no serious adverse events. MRI findings and muscle histology suggested that DPSC treatment downregulated severe inflammation in DMD muscles and demonstrated a milder phenotype after DPSC treatment. DPSC-treated models showed increased recovery in grip-hand strength and improved tetanic force and home cage activity. Interestingly, maintenance of long-term running capability and stabilized cardiac function was also observed in 1-year-old DPSC-treated CXMDJ. Conclusions We developed a novel strategy for the safe and effective transplantation of DPSCs for DMD recovery, which included repeated systemic injection to regulate inflammation at a young age. This is the first report on the efficacy of a systemic DPSC treatment, from which we can propose that DPSCs may play an important role in delaying the DMD disease phenotype.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan. .,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Laboratory of Experimental Animal Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Posadas Herrera Guillermo
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Hiromi Hayashita-Kinoh
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Yasunobu Maruoka
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | | | - Koichi Kimura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of General Medicine, The Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Takashi Okada
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan. .,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan.
| |
Collapse
|
17
|
Sheikh O, Yokota T. Developing DMD therapeutics: a review of the effectiveness of small molecules, stop-codon readthrough, dystrophin gene replacement, and exon-skipping therapies. Expert Opin Investig Drugs 2021; 30:167-176. [PMID: 33393390 DOI: 10.1080/13543784.2021.1868434] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder caused by mutations in the dystrophin (DMD) gene. Most patients die from respiratory failure or cardiomyopathy. There are significant unmet needs for treatments for DMD as the standard of care is principally limited to symptom relief through treatments including steroids. AREAS COVERED This review summarizes safety and efficacy in promising areas of DMD therapeutics - small molecules, stop codon readthrough, gene replacement, and exon skipping - under clinical examination from 2015-2020 as demonstrated in the NIH Clinical Trials and PubMed search engines. EXPERT OPINION Currently, steroids persist as the most accessible medicine for DMD. Stop-codon readthrough, gene replacement, and exon-skipping therapies all aim to restore dystrophin expression. Of these strategies, gene replacement therapy has recently gained momentum while exon-skipping retains great traction. The FDA approval of three exon-skipping antisense oligonucleotides illustrate this regulatory momentum, though the effectiveness and sequence design of eteplirsen remain controversial. Cell-penetrating peptides promise to more efficaciously treat DMD-related cardiomyopathy.The recent success of antisense therapies, however, poses major regulatory challenges. To fully realize the benefits of exon-skipping, including cocktail oligonucleotide-mediated multiple exon-skipping and oligonucleotide drugs for very rare mutations, regulatory challenges need to be addressed in coordination with scientific advances.
Collapse
Affiliation(s)
- Omar Sheikh
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta , Edmonton, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta , Edmonton, Canada
| |
Collapse
|
18
|
Kourakis S, Timpani CA, de Haan JB, Gueven N, Fischer D, Rybalka E. Targeting Nrf2 for the treatment of Duchenne Muscular Dystrophy. Redox Biol 2021; 38:101803. [PMID: 33246292 PMCID: PMC7695875 DOI: 10.1016/j.redox.2020.101803] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/15/2020] [Indexed: 12/15/2022] Open
Abstract
Imbalances in redox homeostasis can result in oxidative stress, which is implicated in various pathological conditions including the fatal neuromuscular disease Duchenne Muscular Dystrophy (DMD). DMD is a complicated disease, with many druggable targets at the cellular and molecular level including calcium-mediated muscle degeneration; mitochondrial dysfunction; oxidative stress; inflammation; insufficient muscle regeneration and dysregulated protein and organelle maintenance. Previous investigative therapeutics tended to isolate and focus on just one of these targets and, consequently, therapeutic activity has been limited. Nuclear erythroid 2-related factor 2 (Nrf2) is a transcription factor that upregulates many cytoprotective gene products in response to oxidants and other toxic stressors. Unlike other strategies, targeted Nrf2 activation has the potential to simultaneously modulate separate pathological features of DMD to amplify therapeutic benefits. Here, we review the literature providing theoretical context for targeting Nrf2 as a disease modifying treatment against DMD.
Collapse
Affiliation(s)
- Stephanie Kourakis
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia.
| | - Cara A Timpani
- Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia; Australian Institute for Musculoskeletal Science, Victoria University, St Albans, Victoria, Australia.
| | - Judy B de Haan
- Oxidative Stress Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Australia.
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Tasmania, Australia.
| | - Dirk Fischer
- Division of Developmental- and Neuropediatrics, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| | - Emma Rybalka
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia; Institute for Health and Sport, Victoria University, Melbourne, Victoria, Australia; Australian Institute for Musculoskeletal Science, Victoria University, St Albans, Victoria, Australia.
| |
Collapse
|
19
|
Anti-Inflammatory and General Glucocorticoid Physiology in Skeletal Muscles Affected by Duchenne Muscular Dystrophy: Exploration of Steroid-Sparing Agents. Int J Mol Sci 2020; 21:ijms21134596. [PMID: 32605223 PMCID: PMC7369834 DOI: 10.3390/ijms21134596] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/13/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD), the activation of proinflammatory and metabolic cellular pathways in skeletal muscle cells is an inherent characteristic. Synthetic glucocorticoid intake counteracts the majority of these mechanisms. However, glucocorticoids induce burdensome secondary effects, including hypertension, arrhythmias, hyperglycemia, osteoporosis, weight gain, growth delay, skin thinning, cushingoid appearance, and tissue-specific glucocorticoid resistance. Hence, lowering the glucocorticoid dosage could be beneficial for DMD patients. A more profound insight into the major cellular pathways that are stabilized after synthetic glucocorticoid administration in DMD is needed when searching for the molecules able to achieve similar pathway stabilization. This review provides a concise overview of the major anti-inflammatory pathways, as well as the metabolic effects of glucocorticoids in the skeletal muscle affected in DMD. The known drugs able to stabilize these pathways, and which could potentially be combined with glucocorticoid therapy as steroid-sparing agents, are described. This could create new opportunities for testing in DMD animal models and/or clinical trials, possibly leading to smaller glucocorticoids dosage regimens for DMD patients.
Collapse
|
20
|
Idebenone Protects against Acute Murine Colitis via Antioxidant and Anti-Inflammatory Mechanisms. Int J Mol Sci 2020; 21:ijms21020484. [PMID: 31940911 PMCID: PMC7013829 DOI: 10.3390/ijms21020484] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Oxidative stress is a key player of the inflammatory cascade responsible for the initiation of ulcerative colitis (UC). Although the short chain quinone idebenone is considered a potent antioxidant and a mitochondrial electron donor, emerging evidence suggests that idebenone also displays anti-inflammatory activity. This study evaluated the impact of idebenone in the widely used dextran sodium sulphate (DSS)-induced mouse model of acute colitis. Acute colitis was induced in C57BL/6J mice via continuous exposure to 2.5% DSS over 7 days. Idebenone was co-administered orally at a dose of 200 mg/kg body weight. Idebenone significantly prevented body weight loss and improved the disease activity index (DAI), colon length, and histopathological score. Consistent with its reported antioxidant function, idebenone significantly reduced the colonic levels of malondialdehyde (MDA) and nitric oxide (NO), and increased the expression of the redox factor NAD(P)H (nicotinamide adenine dinucleotide phosphate) dehydrogenase quinone-1 (NQO-1) in DSS-exposed mice. Immunohistochemistry revealed a significantly increased expression of tight junction proteins, which protect and maintain paracellular intestinal permeability. In support of an anti-inflammatory activity, idebenone significantly attenuated the elevated levels of pro-inflammatory cytokines in colon tissue. These results suggest that idebenone could represent a promising therapeutic strategy to interfere with disease pathology in UC by simultaneously inducing antioxidative and anti-inflammatory pathways.
Collapse
|
21
|
Vitiello L, Tibaudo L, Pegoraro E, Bello L, Canton M. Teaching an Old Molecule New Tricks: Drug Repositioning for Duchenne Muscular Dystrophy. Int J Mol Sci 2019; 20:E6053. [PMID: 31801292 PMCID: PMC6929176 DOI: 10.3390/ijms20236053] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 02/07/2023] Open
Abstract
: Duchenne muscular dystrophy (DMD) is one of the most severe forms of inherited muscular dystrophies. The disease is caused by the lack of dystrophin, a structurally essential protein; hence, a definitive cure would necessarily have to pass through some form of gene and/or cell therapy. Cell- and genetic-based therapeutics for DMD have been explored since the 1990s and recently, two of the latter have been approved for clinical use, but their efficacy is still very low. In parallel, there have been great ongoing efforts aimed at targeting the downstream pathogenic effects of dystrophin deficiency using classical pharmacological approaches, with synthetic or biological molecules. However, as it is always the case with rare diseases, R&D costs for new drugs can represent a major hurdle for researchers and patients alike. This problem can be greatly alleviated by experimenting the use of molecules that had originally been developed for different conditions, a process known as drug repurposing or drug repositioning. In this review, we will describe the state of the art of such an approach for DMD, both in the context of clinical trials and pre-clinical studies.
Collapse
Affiliation(s)
- Libero Vitiello
- Department of Biology, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy;
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
| | - Lucia Tibaudo
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
- Department of Biomedical Sciences, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Via Giustiniani, 5-35128 Padova, Italy;
| | - Luca Bello
- Department of Neurosciences, University of Padova, Via Giustiniani, 5-35128 Padova, Italy;
| | - Marcella Canton
- Interuniversity Institute of Myology (IIM), Administrative headquarters University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy;
- Department of Biomedical Sciences, University of Padova, via U. Bassi 58/B, 35131 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza-IRP, Corso Stati Uniti, 4, 35127 Padova, Italy
| |
Collapse
|
22
|
Servais L, Straathof CSM, Schara U, Klein A, Leinonen M, Hasham S, Meier T, De Waele L, Gordish-Dressman H, McDonald CM, Mayer OH, Voit T, Mercuri E, Buyse GM. Long-term data with idebenone on respiratory function outcomes in patients with Duchenne muscular dystrophy. Neuromuscul Disord 2019; 30:5-16. [PMID: 31813614 DOI: 10.1016/j.nmd.2019.10.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 11/25/2022]
Abstract
Decline in respiratory function in patients with DMD starts during early teenage years and leads to early morbidity and mortality. Published evidence of efficacy for idebenone on respiratory function outcomes is currently limited to 12 months of follow-up time. Here we report data collected as retrospective cohort study (SYROS) from 18 DMD patients not using glucocorticoids who were treated with idebenone (900 mg/day) under Expanded Access Programs (EAPs). The objective was to assess the long-term respiratory function evolution for periods On-Idebenone compared to periods Off-Idebenone in the same patients. The mean idebenone exposure in the EAPs was 4.2 (range 2.4-6.1) years. The primary endpoint was the annual change in forced vital capacity percent of predicted (FVC%p) compared between Off-Idebenone and On-Idebenone periods. The annual rate of decline in FVC%p was reduced by approximately 50% from -7.4% (95% CI: -9.1, -5.8) for the Off-Idebenone periods to -3.8% (95% CI: -4.8, -2.8) for the On-Idebenone periods (N = 11). Similarly, annual change in peak expiratory flow percent of predicted (PEF%p) was -5.9% (95% CI: -8.0, -3.9) for the Off-Idebenone periods (N = 9) and reduced to -1.9% (95% CI: -3.2, -0.7) for the On-Idebenone periods during the EAPs. The reduced rates of decline in FVC%p and PEF%p were maintained for several years with possible beneficial effects on the rate of bronchopulmonary adverse events, time to 10% decline in FVC%p and risk of hospitalization due to respiratory cause. These long-term data provide Class IV evidence to further support the disease modifying treatment effect of idebenone previously observed in randomized, controlled trials.
Collapse
Affiliation(s)
- Laurent Servais
- Centre de Référence Neuromusculaire, CHU Liège, Liège, Belgium
| | - Chiara S M Straathof
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ulrike Schara
- Universitäts-Klinikum Essen, Zentrum für Kinderheilkunde/ Sozialpädiatrisches Zentrum, Essen, Germany
| | - Andrea Klein
- Universität-Kinderspital beider Basel (UKBB) and Inselspital Bern, Neuropädiatrie, Basel and Bern, Switzerland
| | | | | | | | - Liesbeth De Waele
- Pediatric Neurology, University Hospitals Leuven, Herestraat 49, B - 3000 Leuven, Belgium
| | | | | | - Oscar H Mayer
- The Children's Hospital of Philadelphia, Philadelphia, USA
| | - Thomas Voit
- UCL Great Ormond Street Institute of Child Health, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Hospital Trust, London, UK
| | - Eugenio Mercuri
- Paediatric Neurology Unit, Catholic University, Rome, Italy; Centro Nemo, Fodazione Policlinico Gemelli IRCCS, Rome Italy
| | - Gunnar M Buyse
- Pediatric Neurology, University Hospitals Leuven, Herestraat 49, B - 3000 Leuven, Belgium.
| | | |
Collapse
|
23
|
Hughes MC, Ramos SV, Turnbull PC, Rebalka IA, Cao A, Monaco CM, Varah NE, Edgett BA, Huber JS, Tadi P, Delfinis LJ, Schlattner U, Simpson JA, Hawke TJ, Perry CG. Early myopathy in Duchenne muscular dystrophy is associated with elevated mitochondrial H 2 O 2 emission during impaired oxidative phosphorylation. J Cachexia Sarcopenia Muscle 2019; 10:643-661. [PMID: 30938481 PMCID: PMC6596403 DOI: 10.1002/jcsm.12405] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/13/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle wasting and weakness in Duchenne muscular dystrophy (DMD) causes severe locomotor limitations and early death due in part to respiratory muscle failure. Given that current clinical practice focuses on treating secondary complications in this genetic disease, there is a clear need to identify additional contributions in the aetiology of this myopathy for knowledge-guided therapy development. Here, we address the unresolved question of whether the complex impairments observed in DMD are linked to elevated mitochondrial H2 O2 emission in conjunction with impaired oxidative phosphorylation. This study performed a systematic evaluation of the nature and degree of mitochondrial-derived H2 O2 emission and mitochondrial oxidative dysfunction in a mouse model of DMD by designing in vitro bioenergetic assessments that attempt to mimic in vivo conditions known to be critical for the regulation of mitochondrial bioenergetics. METHODS Mitochondrial bioenergetics were compared with functional and histopathological indices of myopathy early in DMD (4 weeks) in D2.B10-DMDmdx /2J mice (D2.mdx)-a model that demonstrates severe muscle weakness. Adenosine diphosphate's (ADP's) central effect of attenuating H2 O2 emission while stimulating respiration was compared under two models of mitochondrial-cytoplasmic phosphate exchange (creatine independent and dependent) in muscles that stained positive for membrane damage (diaphragm, quadriceps, and white gastrocnemius). RESULTS Pathway-specific analyses revealed that Complex I-supported maximal H2 O2 emission was elevated concurrent with a reduced ability of ADP to attenuate emission during respiration in all three muscles (mH2 O2 : +17 to +197% in D2.mdx vs. wild type). This was associated with an impaired ability of ADP to stimulate respiration at sub-maximal and maximal kinetics (-17 to -72% in D2.mdx vs. wild type), as well as a loss of creatine-dependent mitochondrial phosphate shuttling in diaphragm and quadriceps. These changes largely occurred independent of mitochondrial density or abundance of respiratory chain complexes, except for quadriceps. This muscle was also the only one exhibiting decreased calcium retention capacity, which indicates increased sensitivity to calcium-induced permeability transition pore opening. Increased H2 O2 emission was accompanied by a compensatory increase in total glutathione, while oxidative stress markers were unchanged. Mitochondrial bioenergetic dysfunctions were associated with induction of mitochondrial-linked caspase 9, necrosis, and markers of atrophy in some muscles as well as reduced hindlimb torque and reduced respiratory muscle function. CONCLUSIONS These results provide evidence that Complex I dysfunction and loss of central respiratory control by ADP and creatine cause elevated oxidant generation during impaired oxidative phosphorylation. These dysfunctions may contribute to early stage disease pathophysiology and support the growing notion that mitochondria are a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Meghan C. Hughes
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Sofhia V. Ramos
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Patrick C. Turnbull
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Irena A. Rebalka
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Andrew Cao
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Cynthia M.F. Monaco
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Nina E. Varah
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Brittany A. Edgett
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Jason S. Huber
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Peyman Tadi
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Luca J. Delfinis
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - U. Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy)University Grenoble AlpesGrenobleFrance
| | - Jeremy A. Simpson
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Thomas J. Hawke
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Christopher G.R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| |
Collapse
|
24
|
|
25
|
Hughes MC, Ramos SV, Turnbull PC, Edgett BA, Huber JS, Polidovitch N, Schlattner U, Backx PH, Simpson JA, Perry CGR. Impairments in left ventricular mitochondrial bioenergetics precede overt cardiac dysfunction and remodelling in Duchenne muscular dystrophy. J Physiol 2019; 598:1377-1392. [PMID: 30674086 DOI: 10.1113/jp277306] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 01/22/2019] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Ninety-eight per cent of patients with Duchenne muscular dystrophy (DMD) develop cardiomyopathy, with 40% developing heart failure. While increased propensity for mitochondrial induction of cell death has been observed in left ventricle, it remains unknown whether this is linked to impaired mitochondrial respiratory control and elevated H2 O2 emission prior to the onset of cardiomyopathy. Classic mouse models of DMD demonstrate hyper-regeneration in skeletal muscle which may mask mitochondrial abnormalities. Using a model with less regenerative capacity that is more akin to DMD patients, we observed elevated left ventricular mitochondrial H2 O2 and impaired oxidative phosphorylation in the absence of cardiac remodelling or overt cardiac dysfunction at 4 weeks. These impairments were associated with dysfunctions at complex I, governance by ADP and creatine-dependent phosphate shuttling, which results in a less efficient response to energy demands. Mitochondria may be a therapeutic target for the treatment of cardiomyopathy in DMD. ABSTRACT In Duchenne muscular dystrophy (DMD), mitochondrial dysfunction is predicted as a response to numerous cellular stressors, yet the contribution of mitochondria to the onset of cardiomyopathy remains unknown. To resolve this uncertainty, we designed in vitro assessments of mitochondrial bioenergetics to model mitochondrial control parameters that influence cardiac function. Both left ventricular mitochondrial responsiveness to the central bioenergetic controller ADP and the ability of creatine to facilitate mitochondrial-cytoplasmic phosphate shuttling were assessed. These measurements were performed in D2.B10-DMDmdx /2J mice - a model that demonstrates skeletal muscle atrophy and weakness due to limited regenerative capacities and cardiomyopathy more akin to people with DMD than classic models. At 4 weeks of age, there was no evidence of cardiac remodelling or cardiac dysfunction despite impairments in ADP-stimulated respiration and ADP attenuation of H2 O2 emission. These impairments were seen at both submaximal and maximal ADP concentrations despite no reductions in mitochondrial content markers. The ability of creatine to enhance ADP's control of mitochondrial bioenergetics was also impaired, suggesting an impairment in mitochondrial creatine kinase-dependent phosphate shuttling. Susceptibly to permeability transition pore opening and the subsequent activation of cell death pathways remained unchanged. Mitochondrial H2 O2 emission was elevated despite no change in markers of irreversible oxidative damage, suggesting alternative redox signalling mechanisms should be explored. These findings demonstrate that selective mitochondrial dysfunction precedes the onset of overt cardiomyopathy in D2.mdx mice, suggesting that improving mitochondrial bioenergetics by restoring ADP, creatine-dependent phosphate shuttling and complex I should be considered for treating DMD patients.
Collapse
Affiliation(s)
- Meghan C Hughes
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Sofhia V Ramos
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Patrick C Turnbull
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Brittany A Edgett
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada.,Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada.,IMPART Team Canada Investigator Network, Saint John, New Brunswick, Canada
| | - Jason S Huber
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada
| | - Nazari Polidovitch
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy), University Grenoble Alpes, Grenoble, France
| | - Peter H Backx
- Department of Biology and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences and Cardiovascular Research Group, University of Guelph, Guelph, ON, Canada.,IMPART Team Canada Investigator Network, Saint John, New Brunswick, Canada
| | - Christopher G R Perry
- School of Kinesiology and Health Science and the Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
26
|
At the Crossroads of Clinical and Preclinical Research for Muscular Dystrophy-Are We Closer to Effective Treatment for Patients? Int J Mol Sci 2018; 19:ijms19051490. [PMID: 29772730 PMCID: PMC5983724 DOI: 10.3390/ijms19051490] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/13/2022] Open
Abstract
Among diseases affecting skeletal muscle, muscular dystrophy is one of the most devastating and complex disorders. The term ‘muscular dystrophy’ refers to a heterogeneous group of genetic diseases associated with a primary muscle defect that leads to progressive muscle wasting and consequent loss of muscle function. Muscular dystrophies are accompanied by numerous clinical complications and abnormalities in other tissues that cause extreme discomfort in everyday life. The fact that muscular dystrophy often takes its toll on babies and small children, and that many patients die at a young age, adds to the cruel character of the disease. Clinicians all over the world are facing the same problem: they have no therapy to offer except for symptom-relieving interventions. Patients, their families, but also clinicians, are in urgent need of an effective cure. Despite advances in genetics, increased understanding of molecular mechanisms underlying muscle disease, despite a sweeping range of successful preclinical strategies and relative progress of their implementation in the clinic, therapy for patients is currently out of reach. Only a greater comprehension of disease mechanisms, new preclinical studies, development of novel technologies, and tight collaboration between scientists and physicians can help improve clinical treatment. Fortunately, inventiveness in research is rapidly extending the limits and setting new standards for treatment design. This review provides a synopsis of muscular dystrophy and considers the steps of preclinical and clinical research that are taking the muscular dystrophy community towards the fundamental goal of combating the traumatic disease.
Collapse
|
27
|
Tempol Supplementation Restores Diaphragm Force and Metabolic Enzyme Activities in mdx Mice. Antioxidants (Basel) 2017; 6:antiox6040101. [PMID: 29210997 PMCID: PMC5745511 DOI: 10.3390/antiox6040101] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/19/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by striated muscle weakness, cardiomyopathy, and respiratory failure. Since oxidative stress is recognized as a secondary pathology in DMD, the efficacy of antioxidant intervention, using the superoxide scavenger tempol, was examined on functional and biochemical status of dystrophin-deficient diaphragm muscle. Diaphragm muscle function was assessed, ex vivo, in adult male wild-type and dystrophin-deficient mdx mice, with and without a 14-day antioxidant intervention. The enzymatic activities of muscle citrate synthase, phosphofructokinase, and lactate dehydrogenase were assessed using spectrophotometric assays. Dystrophic diaphragm displayed mechanical dysfunction and altered biochemical status. Chronic tempol supplementation in the drinking water increased diaphragm functional capacity and citrate synthase and lactate dehydrogenase enzymatic activities, restoring all values to wild-type levels. Chronic supplementation with tempol recovers force-generating capacity and metabolic enzyme activity in mdx diaphragm. These findings may have relevance in the search for therapeutic strategies in neuromuscular disease.
Collapse
|
28
|
Insights into the Pathogenic Secondary Symptoms Caused by the Primary Loss of Dystrophin. J Funct Morphol Kinesiol 2017. [DOI: 10.3390/jfmk2040044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
29
|
Salehi F, Zeinaloo A, Riasi HR, Shamloo AS. Effectiveness of Coenzyme Q10 on echocardiographic parameters of patients with Duchenne muscular dystrophy. Electron Physician 2017; 9:3896-3904. [PMID: 28461862 PMCID: PMC5407220 DOI: 10.19082/3896] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 12/23/2016] [Indexed: 02/02/2023] Open
Abstract
Background Myocardial damage is a common complication in patients with Duchenne muscular dystrophy (DMD) that occurs due to myocardial replacement by fat and fibrosis. In recent years, efforts have been made toward finding new pharmacological agents with fewer complications which can be used as prophylactic before the symptoms. Coenzyme Q10 plays a central role in production of bioenergy in heart muscle and antioxidant in reperfusion condition of myocardial damaged muscle and leads to membrane stability and prevents cell death. Objective This study aimed at comparing the Effectiveness of coenzyme Q10 on echocardiographic parameters of pediatric patients with Duchenne muscular dystrophy. Methods This randomized clinical trial study (RCT) was carried out on 25 pediatric patients with pre-diagnosed DMD who attended the Children’s Medical Center (CMC), Tehran, Iran from February 2013 to 2015. The patients were randomly divided into two groups. Group-1; (n=12) was treated with coenzyme Q10 for six months and group-2 ;(n=13) received placebo for the same time. The primary aim was to compare the myocardial performance index (MPI), between the two groups at the end of six months. Data were analyzed by SPSS software (ver-16) and using T-Test. Results Twenty-five patients under study were divided into two groups of (Q10=12) and (placebo=13). Mean ages were 8.9±1.7 and 8.6±1.4 in Q10 and placebo groups (P=0.66). No significant difference was detected in MPI at all three views of mitral and tricuspid and septum respectively in two groups after the end of treatment (0.41±0.13, and 0.43±0.6; P=0.59), (0.45±0.12, and 0.46±0.1; P=0.05), and (0.45±0.06, and 0.45±0.1; P=0.31). Conclusion According to the results obtained from this study, coenzyme Q10 had no significant effect on improving the performance of echocardiographic parameters in patients with DMD. Trial registration The trial is registered at the Iranian Clinical Trial Registry (IRCT.ir) with the IRCT identification number IRCT2015070223018N1. Funding This research has been financially supported by the Research Council of Tehran University of Medical Sciences.
Collapse
Affiliation(s)
- Forod Salehi
- M.D., Pediatric Cardiologist, Assistant Professor, Birjand CardioVascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Aliakbar Zeinaloo
- M.D., Pediatric Cardiologist, Professor, Pediatric Cardiology Department, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Riasi
- M.D., Neurologist, Associate Professor, Birjand University of Medical Sciences, Birjand, Iran
| | | |
Collapse
|
30
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
31
|
Nitahara-Kasahara Y, Takeda S, Okada T. Inflammatory predisposition predicts disease phenotypes in muscular dystrophy. Inflamm Regen 2016; 36:14. [PMID: 29259687 PMCID: PMC5725653 DOI: 10.1186/s41232-016-0019-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 06/01/2016] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy is an incurable genetic disease that presents with skeletal muscle weakness and chronic inflammation and is associated with early mortality. Indeed, immune cell infiltration into the skeletal muscle is a notable feature of the disease pathophysiology and is strongly associated with disease severity. Interleukin (IL)-10 regulates inflammatory immune responses by reducing both M1 macrophage activation and the production of pro-inflammatory cytokines, thereby promoting the activation of the M2 macrophage phenotype. We previously reported that genetic ablation of IL-10 in dystrophic mice resulted in more severe phenotypes, in regard to heart and respiratory function, as evidenced by increased macrophage infiltration, high levels of inflammatory factors in the muscle, and progressive cardiorespiratory dysfunction. These data therefore indicate that IL-10 comprises an essential immune-modulator within dystrophic muscles. In this review, we highlight the pivotal role of the immune system in the pathogenesis of muscular dystrophy and discuss how an increased understanding of the pathogenesis of this disease may lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo-ku Tokyo, Japan.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira Tokyo, Japan
| | - Takashi Okada
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Bunkyo-ku Tokyo, Japan.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira Tokyo, Japan
| |
Collapse
|
32
|
Miyatake S, Shimizu-Motohashi Y, Takeda S, Aoki Y. Anti-inflammatory drugs for Duchenne muscular dystrophy: focus on skeletal muscle-releasing factors. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2745-58. [PMID: 27621596 PMCID: PMC5012616 DOI: 10.2147/dddt.s110163] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD), an incurable and a progressive muscle wasting disease, is caused by the absence of dystrophin protein, leading to recurrent muscle fiber damage during contraction. The inflammatory response to fiber damage is a compelling candidate mechanism for disease exacerbation. The only established pharmacological treatment for DMD is corticosteroids to suppress muscle inflammation, however this treatment is limited by its insufficient therapeutic efficacy and considerable side effects. Recent reports show the therapeutic potential of inhibiting or enhancing pro- or anti-inflammatory factors released from DMD skeletal muscles, resulting in significant recovery from muscle atrophy and dysfunction. We discuss and review the recent findings of DMD inflammation and opportunities for drug development targeting specific releasing factors from skeletal muscles. It has been speculated that nonsteroidal anti-inflammatory drugs targeting specific inflammatory factors are more effective and have less side effects for DMD compared with steroidal drugs. For example, calcium channels, reactive oxygen species, and nuclear factor-κB signaling factors are the most promising targets as master regulators of inflammatory response in DMD skeletal muscles. If they are combined with an oligonucleotide-based exon skipping therapy to restore dystrophin expression, the anti-inflammatory drug therapies may address the present therapeutic limitation of low efficiency for DMD.
Collapse
Affiliation(s)
- Shouta Miyatake
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuko Shimizu-Motohashi
- Department of Child Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| |
Collapse
|
33
|
Abstract
Cardiomyopathy is an inherited or acquired disease of the myocardium, which can result in severe ventricular dysfunction. Mitochondrial dysfunction is involved in the pathological process of cardiomyopathy. Many dysfunctions in cardiac mitochondria are consequences of mutations in nuclear or mitochondrial DNA followed by alterations in transcriptional regulation, mitochondrial protein function, and mitochondrial dynamics and energetics, presenting with associated multisystem mitochondrial disorders. To ensure correct diagnosis and optimal management of mitochondrial dysfunction in cardiomyopathy caused by multiple pathogenesis, multidisciplinary approaches are required, and to integrate between clinical and basic sciences, ideal translational models are needed. In this review, we will focus on experimental models to provide insights into basic mitochondrial physiology and detailed underlying mechanisms of cardiomyopathy and current mitochondria-targeted therapies for cardiomyopathy. [BMB Reports 2015; 48(10): 541-548]
Collapse
Affiliation(s)
- Youn Wook Chung
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seok-Min Kang
- Yonsei Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 03722; Cardiology Division, Severance Cardiovascular Hospital, Seoul 03722; Severance Integrative Research Institute for Cerebral and Cardiovascular Diseases (SIRIC), Yonsei University Health System, Seoul 03722, Korea
| |
Collapse
|
34
|
Moulin M, Ferreiro A. Muscle redox disturbances and oxidative stress as pathomechanisms and therapeutic targets in early-onset myopathies. Semin Cell Dev Biol 2016; 64:213-223. [PMID: 27531051 DOI: 10.1016/j.semcdb.2016.08.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/08/2016] [Indexed: 01/07/2023]
Abstract
Because of their contractile activity and their high oxygen consumption and metabolic rate, skeletal muscles continually produce moderate levels of reactive oxygen and nitrogen species (ROS/RNS), which increase during exercise and are buffered by multiple antioxidant systems to maintain redox homeostasis. Imbalance between ROS/RNS production and elimination results in oxidative stress (OxS), which has been implicated in ageing and in numerous human diseases, including cancer, diabetes or age-related muscle loss (sarcopenia). The study of redox homeostasis in muscle was hindered by its lability, by the many factors influencing technical OxS measures and by ROS/RNS important roles in signaling pathways and adaptative responses to muscle contraction and effort, which make it difficult to define a threshold between physiological signaling and pathological conditions. In the last years, new tools have been developed that facilitate the study of these key mechanisms, and deregulation of redox homeostasis has emerged as a key pathogenic mechanism and potential therapeutic target in muscle conditions. This is in particular the case for early-onset myopathies, genetic muscle diseases which present from birth or early childhood with muscle weakness interfering with ambulation and often with cardiac or respiratory failure leading to premature death. Inherited defects of the reductase selenoprotein N in SEPN1-related myopathy leads to chronic OxS of monogenic origin as a primary disease pathomechanism. In myopathies associated with mutations of the genes encoding the calcium channel RyR1, the extracellular matrix protein collagen VI or the sarcolemmal protein dystrophin (Duchenne Muscular Dystrophy), OxS has been identified as a relevant secondary pathophysiological mechanism. OxS being drug-targetable, it represents an interesting therapeutic target for these incurable conditions, and following preclinical correction of the cell or animal model phenotype, the first clinical trials with the antioxidants N-acetylcysteine (SEPN1- and RYR1-related myopathies) or epigallocatechin-gallate (DMD) have been launched recently. In this review, we provide an overview of the mechanisms involved in redox regulation in skeletal muscle, the technical tools available to measure redox homeostasis in muscle cells, the bases of OxS as a primary or secondary pathomechanism in early-onset myopathies and the innovative clinical trials with antioxidants which are currently in progress for these so-far untreatable infantile muscle diseases. Progress in our knowledge of redox homeostasis defects in these rare muscle conditions may be useful as a model paradigm to understand and treat other conditions in which OxS is involved, including prevalent conditions with major socioeconomic impact such as insulin resistance, cachexia, obesity, sarcopenia or ageing.
Collapse
Affiliation(s)
- Maryline Moulin
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250, Paris Cedex 13, France.
| | - Ana Ferreiro
- Pathophysiology of Striated Muscles Laboratory, Unit of Functional and Adaptive Biology (BFA), University Paris Diderot, Sorbonne Paris Cité, BFA, UMR CNRS 8251, 75250, Paris Cedex 13, France; AP-HP, Centre de Référence Maladies Neuromusculaires Paris-Est, Groupe Hospitalier Pitié-Salpêtrière, 75013, Paris, France.
| |
Collapse
|
35
|
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy in childhood. It is caused by mutations of the DMD gene, leading to progressive muscle weakness, loss of independent ambulation by early teens, and premature death due to cardiorespiratory complications. The diagnosis can usually be made after careful review of the history and examination of affected boys presenting with developmental delay, proximal weakness, and elevated serum creatine kinase, plus confirmation by muscle biopsy or genetic testing. Precise characterization of the DMD mutation is important for genetic counseling and individualized treatment. Current standard of care includes the use of corticosteroids to prolong ambulation and to delay the onset of secondary complications. Early use of cardioprotective agents, noninvasive positive pressure ventilation, and other supportive strategies has improved the life expectancy and health-related quality of life for many young adults with DMD. New emerging treatment includes viral-mediated microdystrophin gene replacement, exon skipping to restore the reading frame, and nonsense suppression therapy to allow translation and production of a modified dystrophin protein. Other potential therapeutic targets involve upregulation of compensatory proteins, reduction of the inflammatory cascade, and enhancement of muscle regeneration. So far, data from DMD clinical trials have shown limited success in delaying disease progression; unforeseen obstacles included immune response against the generated mini-dystrophin, inconsistent evidence of dystrophin production in muscle biopsies, and failure to demonstrate a significant improvement in the primary outcome measure, as defined by the 6-minute walk test in some studies. The long-term safety and efficacy of emerging treatments will depend on the selection of appropriate clinical end points and sensitive biomarkers to detect meaningful changes in disease progression. Correction of the underlying mutations using new gene-editing technologies and corticosteroid analogs with better safety profiles offers renewed hope for many individuals with DMD and their families.
Collapse
Affiliation(s)
- Jean K Mah
- Department of Pediatrics and Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
36
|
Oxidative Stress-Mediated Skeletal Muscle Degeneration: Molecules, Mechanisms, and Therapies. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:6842568. [PMID: 26798425 PMCID: PMC4700198 DOI: 10.1155/2016/6842568] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/08/2015] [Accepted: 10/08/2015] [Indexed: 11/25/2022]
Abstract
Oxidative stress is a loss of balance between the production of reactive oxygen species during cellular metabolism and the mechanisms that clear these species to maintain cellular redox homeostasis. Increased oxidative stress has been associated with muscular dystrophy, and many studies have proposed mechanisms that bridge these two pathological conditions at the molecular level. In this review, the evidence indicating a causal role of oxidative stress in the pathogenesis of various muscular dystrophies is revisited. In particular, the mediation of cellular redox status in dystrophic muscle by NF-κB pathway, autophagy, telomere shortening, and epigenetic regulation are discussed. Lastly, the current stance of targeting these pathways using antioxidant therapies in preclinical and clinical trials is examined.
Collapse
|
37
|
|
38
|
Rybalka E, Timpani CA, Stathis CG, Hayes A, Cooke MB. Metabogenic and Nutriceutical Approaches to Address Energy Dysregulation and Skeletal Muscle Wasting in Duchenne Muscular Dystrophy. Nutrients 2015; 7:9734-67. [PMID: 26703720 PMCID: PMC4690050 DOI: 10.3390/nu7125498] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/29/2015] [Accepted: 11/13/2015] [Indexed: 12/21/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal genetic muscle wasting disease with no current cure. A prominent, yet poorly treated feature of dystrophic muscle is the dysregulation of energy homeostasis which may be associated with intrinsic defects in key energy systems and promote muscle wasting. As such, supplementative nutriceuticals that target and augment the bioenergetical expansion of the metabolic pathways involved in cellular energy production have been widely investigated for their therapeutic efficacy in the treatment of DMD. We describe the metabolic nuances of dystrophin-deficient skeletal muscle and review the potential of various metabogenic and nutriceutical compounds to ameliorate the pathological and clinical progression of the disease.
Collapse
Affiliation(s)
- Emma Rybalka
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Cara A Timpani
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
| | - Christos G Stathis
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Alan Hayes
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Matthew B Cooke
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| |
Collapse
|
39
|
Peverelli L, Testolin S, Villa L, D'Amico A, Petrini S, Favero C, Magri F, Morandi L, Mora M, Mongini T, Bertini E, Sciacco M, Comi GP, Moggio M. Histologic muscular history in steroid-treated and untreated patients with Duchenne dystrophy. Neurology 2015; 85:1886-93. [PMID: 26497992 DOI: 10.1212/wnl.0000000000002147] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is a lethal disease. The outcome measures used in numerous therapeutic trials include skeletal muscle biopsy. We studied the natural history of DMD from the standpoint of muscle histology with the aim of providing a reproducible tool for use in evaluating and comparing any histologic changes occurring in patients with DMD undergoing treatment and hence be able to determine how therapy modulates the histologic evolution of the disease. METHODS Three independent operators analyzed 56 muscle biopsies from 40 patients not treated with steroids, aged 1 to 10 years and 16 individuals treated with steroids, aged 7 to 10 years. We analyzed morphologic measures, normalized every measure for the average number of fibers observed for each year of age, and calculated intraclass correlation coefficients. RESULTS The average proportion of connective tissue in patients not treated with steroids was 16.98% from ages 1 to 6 years and 30% from ages 7 to 10 years (p < 0.0001). The average proportion in patients treated with steroids was 24.90%. Muscle fiber area mirrored that of connective tissue in both groups. CONCLUSIONS Having provided a reproducible tool for evaluation and comparison of histologic changes occurring in patients undergoing clinical trials, it was observed that at ages 6 to 7 years, fibrotic tissue rapidly peaks to 29.85%; this is a crucial moment when muscle tissue loses its self-regeneration ability, veering toward fibrotic degeneration. These data should be considered when deciding the most suitable time to begin therapy.
Collapse
Affiliation(s)
- Lorenzo Peverelli
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Silvia Testolin
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Luisa Villa
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Adele D'Amico
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Stefania Petrini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Chiara Favero
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Francesca Magri
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Lucia Morandi
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Marina Mora
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Tiziana Mongini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Enrico Bertini
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Monica Sciacco
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Giacomo P Comi
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy
| | - Maurizio Moggio
- From the Neuromuscular and Rare Diseases Unit (L.P., S.T., L.V., M.S., M. Moggio), Department of Neuroscience, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan; Laboratory of Molecular Medicine for Muscular and Neurodegenerative Diseases (A.D.), Research Center, Confocal Microscopy Facility (S.P.), and Department of Neurosciences, Unit of Neuromuscular and Neurodegenerative Disorders (E.B.), Bambino Gesù Children's Hospital, Rome; Center of Molecular and Genetic Epidemiology (C.F.), Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan; Dino Ferrari Center (F.M., G.P.C.), Neuroscience Section, Department of Pathophysiology and Transplantation, University of Milan, Neurology Unit, IRCCS Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan; U.O. Neuromuscular Diseases and Neuroimmunology (L.M., M. Mora), Fondazione IRCCS Istituto Neurologico C. Besta, Milan; and Department of Neurosciences Rita Levi Montalcini (T.M.), University of Turin, Italy.
| |
Collapse
|
40
|
LoMauro A, D'Angelo MG, Aliverti A. Assessment and management of respiratory function in patients with Duchenne muscular dystrophy: current and emerging options. Ther Clin Risk Manag 2015; 11:1475-88. [PMID: 26451113 PMCID: PMC4592047 DOI: 10.2147/tcrm.s55889] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked myopathy resulting in progressive weakness and wasting of all the striated muscles including the respiratory muscles. The consequences are loss of ambulation before teen ages, cardiac involvement and breathing difficulties, the main cause of death. A cure for DMD is not currently available. In the last decades the survival of patients with DMD has improved because the natural history of the disease can be changed thanks to a more comprehensive therapeutic approach. This comprises interventions targeted to the manifestations and complications of the disease, particularly in the respiratory care. These include: 1) pharmacological intervention, namely corticosteroids and idebenone that significantly reduce the decline of spirometric parameters; 2) rehabilitative intervention, namely lung volume recruitment techniques that help prevent atelectasis and slows the rate of decline of pulmonary function; 3) scoliosis treatment, namely steroid therapy that is used to reduce muscle inflammation/degeneration and prolong ambulation in order to delay the onset of scoliosis, being an additional contribution to the restrictive lung pattern; 4) cough assisted devices that improve airway clearance thus reducing the risk of pulmonary infections; and 5) non-invasive mechanical ventilation that is essential to treat nocturnal hypoventilation, sleep disordered breathing, and ultimately respiratory failure. Without any intervention death occurs within the first 2 decades, however, thanks to this multidisciplinary therapeutic approach life expectancy of a newborn with DMD nowadays can be significantly prolonged up to his fourth decade. This review is aimed at providing state-of-the-art methods and techniques for the assessment and management of respiratory function in DMD patients.
Collapse
Affiliation(s)
- Antonella LoMauro
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | | | - Andrea Aliverti
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| |
Collapse
|
41
|
Swiderski K, Lynch GS. Therapeutic potential of orphan drugs for the rare skeletal muscle diseases. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1085858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
Arend N, Wertheimer C, Laubichler P, Wolf A, Kampik A, Kernt M. Idebenone Prevents Oxidative Stress, Cell Death and Senescence of Retinal Pigment Epithelium Cells by Stabilizing BAX/Bcl-2 Ratio. Ophthalmologica 2015; 234:73-82. [PMID: 26044821 DOI: 10.1159/000381726] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 03/13/2015] [Indexed: 11/19/2022]
Abstract
PURPOSE Age-related macular degeneration (AMD) is one of the leading causes of blindness. Degeneration of the retinal pigment epithelium (RPE) is pathognomonic for the disease, and oxidative stress plays an important role in the pathogenesis of this disease. This study investigates potential antiapoptotic and cytoprotective effects of idebenone on cultured RPE cells (ARPE-19) under conditions of oxidative stress. METHODS ARPE-19 cells were treated with 1-100 µ<smlcap>M</smlcap> idebenone. Cell viability (MTT assay), induction of intracellular reactive oxygen species (ROS) and histone-associated DNA fragments in mono- and oligonucleosomes, expression of proapoptotic BAX and antiapoptotic Bcl-2 as well as senescence-associated β-galactosidase (SA-β-Gal) activity were investigated under exposure to hydrogen peroxide (H2O2). RESULTS Idebenone concentrations from 1 to 20 µ<smlcap>M</smlcap> showed no toxic effects on ARPE-19 cells. When cells were treated with H2O2, pretreatment with 5, 7.5, 10, and 20 µ<smlcap>M</smlcap> idebenone led to a significant increase in the viability of ARPE-19 cells. In addition, idebenone pretreatment significantly attenuated the induction of SA-β-Gal and intracellular ROS as well as the amount of histone-associated DNA fragments after treatment with H2O2. The reduction of proapoptotic BAX and the elevation of antiapoptotic Bcl-2 under idebenone show that this process is rather mediated by inhibiting H2O2-induced apoptosis, not necrosis. CONCLUSION In this study, idebenone increased survival of ARPE-19 cells and reduced cell death, senescence, and oxidative stress by stabilizing the BAX/Bcl-2 ratio.
Collapse
Affiliation(s)
- Nicole Arend
- Department of Ophthalmology, Ludwig Maximilian University, Munich, Germany
| | | | | | | | | | | |
Collapse
|
43
|
Blat Y, Blat S. Drug Discovery of Therapies for Duchenne Muscular Dystrophy. ACTA ACUST UNITED AC 2015; 20:1189-203. [PMID: 25975656 DOI: 10.1177/1087057115586535] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 04/21/2015] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic, lethal, muscle disorder caused by the loss of the muscle protein, dystrophin, leading to progressive loss of muscle fibers and muscle weakness. Drug discovery efforts targeting DMD have used two main approaches: (1) the restoration of dystrophin expression or the expression of a compensatory protein, and (2) the mitigation of downstream pathological mechanisms, including dysregulated calcium homeostasis, oxidative stress, inflammation, fibrosis, and muscle ischemia. The aim of this review is to introduce the disease, its pathophysiology, and the available research tools to a drug discovery audience. This review will also detail the most promising therapies that are currently being tested in clinical trials or in advanced preclinical models.
Collapse
Affiliation(s)
| | - Shachar Blat
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
Buyse GM, Voit T, Schara U, Straathof CSM, D'Angelo MG, Bernert G, Cuisset JM, Finkel RS, Goemans N, McDonald CM, Rummey C, Meier T. Efficacy of idebenone on respiratory function in patients with Duchenne muscular dystrophy not using glucocorticoids (DELOS): a double-blind randomised placebo-controlled phase 3 trial. Lancet 2015; 385:1748-1757. [PMID: 25907158 DOI: 10.1016/s0140-6736(15)60025-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cardiorespiratory failure is the leading cause of death in Duchenne muscular dystrophy. Based on preclinical and phase 2 evidence, we assessed the efficacy and safety of idebenone in young patients with Duchenne muscular dystrophy who were not taking concomitant glucocorticoids. METHODS In a multicentre phase 3 trial in Belgium, Germany, the Netherlands, Switzerland, France, Sweden, Austria, Italy, Spain, and the USA, patients (age 10-18 years old) with Duchenne muscular dystrophy were randomly assigned in a one-to-one ratio with a central interactive web response system with a permuted block design with four patients per block to receive idebenone (300 mg three times a day) or matching placebo orally for 52 weeks. Study personnel and patients were masked to treatment assignment. The primary endpoint was change in peak expiratory flow (PEF) as percentage predicted (PEF%p) from baseline to week 52, measured with spirometry. Analysis was by intention to treat (ITT) and a modified ITT (mITT), which was prospectively defined to exclude patients with at least 20% difference in the yearly change in PEF%p, measured with hospital-based and weekly home-based spirometry. This study is registered with ClinicalTrials.gov, number NCT01027884. FINDINGS 31 patients in the idebenone group and 33 in the placebo group comprised the ITT population, and 30 and 27 comprised the mITT population. Idebenone significantly attenuated the fall in PEF%p from baseline to week 52 in the mITT (-3·05%p [95% CI -7·08 to 0·97], p=0·134, vs placebo -9·01%p [-13·18 to -4·84], p=0·0001; difference 5·96%p [0·16 to 11·76], p=0·044) and ITT populations (-2·57%p [-6·68 to 1·54], p=0·215, vs -8·84%p [-12·73 to -4·95], p<0·0001; difference 6·27%p [0·61 to 11·93], p=0·031). Idebenone also had a significant effect on PEF (L/min), weekly home-based PEF, FVC, and FEV1. The effect of idebenone on respiratory function outcomes was similar between patients with previous corticosteroid use and steroid-naive patients. Treatment with idebenone was safe and well tolerated with adverse event rates were similar in both groups. Nasopharyngitis and headache were the most common adverse events (idebenone, eight [25%] and six [19%] of 32 patients; placebo, nine [26%] and seven [21%] of 34 patients). Transient and mild diarrhoea was more common in the idebenone group than in the placebo group (eight [25%] vs four [12%] patients). INTERPRETATION Idebenone reduced the loss of respiratory function and represents a new treatment option for patients with Duchenne muscular dystrophy. FUNDING Santhera Pharmaceuticals.
Collapse
Affiliation(s)
| | - Thomas Voit
- Institut de Myologie, Université Pierre et Marie Curie INSERM UMR 974, CNRS FRE 3617, Groupe Hospitalier de la Pitié Salpetrière, Paris, France
| | | | | | - M Grazia D'Angelo
- Istituto di Ricovero e Cura a Carattere Scientifico Eugenio Medea, Lecco, Italy
| | | | | | | | | | - Craig M McDonald
- University of California Davis Medical Center, Sacramento, CA, USA
| | | | | |
Collapse
|
45
|
Passerieux E, Hayot M, Jaussent A, Carnac G, Gouzi F, Pillard F, Picot MC, Böcker K, Hugon G, Pincemail J, Defraigne JO, Verrips T, Mercier J, Laoudj-Chenivesse D. Effects of vitamin C, vitamin E, zinc gluconate, and selenomethionine supplementation on muscle function and oxidative stress biomarkers in patients with facioscapulohumeral dystrophy: a double-blind randomized controlled clinical trial. Free Radic Biol Med 2015; 81:158-69. [PMID: 25246239 DOI: 10.1016/j.freeradbiomed.2014.09.014] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/11/2014] [Accepted: 09/11/2014] [Indexed: 12/29/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disease characterized by progressive weakness and atrophy of specific skeletal muscles. As growing evidence suggests that oxidative stress may contribute to FSHD pathology, antioxidants that might modulate or delay oxidative insults could help in maintaining FSHD muscle function. Our primary objective was to test whether oral administration of vitamin C, vitamin E, zinc gluconate, and selenomethionine could improve the physical performance of patients with FSHD. Adult patients with FSHD (n=53) were enrolled at Montpellier University Hospital (France) in a randomized, double-blind, placebo-controlled pilot clinical trial. Patients were randomly assigned to receive 500 mg vitamin C, 400mg vitamin E, 25mg zinc gluconate and 200 μg selenomethionine (n=26), or matching placebo (n=27) once a day for 17 weeks. Primary outcomes were changes in the two-minute walking test (2-MWT), maximal voluntary contraction, and endurance limit time of the dominant and nondominant quadriceps (MVCQD, MVCQND, TlimQD, and TlimQND, respectively) after 17 weeks of treatment. Secondary outcomes were changes in the antioxidant status and oxidative stress markers. Although 2-MWT, MVCQ, and TlimQ were all significantly improved in the supplemented group at the end of the treatment compared to baseline, only MVCQ and TlimQ variations were significantly different between groups (MVCQD: P=0.011; MVCQND: P=0.004; TlimQD: P=0.028; TlimQND: P=0.011). Similarly, the vitamin C (P<0.001), vitamin E as α-tocopherol (P<0.001), vitamin C/vitamin E ratio (P=0.017), vitamin E γ/α ratio (P=0.022) and lipid peroxides (P<0.001) variations were significantly different between groups. In conclusion, vitamin E, vitamin C, zinc, and selenium supplementation has no significant effect on the 2-MWT, but improves MVCQ and TlimQ of both quadriceps by enhancing the antioxidant defenses and reducing oxidative stress. This trial was registered at clinicaltrials.gov (number: NCT01596803).
Collapse
Affiliation(s)
- Emilie Passerieux
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Maurice Hayot
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Audrey Jaussent
- Department of Biostatistics and Epidemiology, University Hospital of Montpellier, Montpellier, France
| | - Gilles Carnac
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Fares Gouzi
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Fabien Pillard
- Department of Respiratory Exploration and Department of Sports Medicine, Larrey University Hospital, Toulouse CEDEX, France
| | - Marie-Christine Picot
- Department of Biostatistics and Epidemiology, University Hospital, Montpellier, France and CIC 1001-INSERM
| | - Koen Böcker
- Alan Turing Institute Almere, The Netherlands
| | - Gerald Hugon
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Joel Pincemail
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Jean O Defraigne
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Theo Verrips
- Utrecht University, Department of Biology, The Netherlands
| | - Jacques Mercier
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Dalila Laoudj-Chenivesse
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France.
| |
Collapse
|
46
|
Betts CA, Saleh AF, Carr CA, Hammond SM, Coenen-Stass AML, Godfrey C, McClorey G, Varela MA, Roberts TC, Clarke K, Gait MJ, Wood MJA. Prevention of exercised induced cardiomyopathy following Pip-PMO treatment in dystrophic mdx mice. Sci Rep 2015; 5:8986. [PMID: 25758104 PMCID: PMC4355666 DOI: 10.1038/srep08986] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/03/2015] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal neuromuscular disorder caused by mutations in the Dmd gene. In addition to skeletal muscle wasting, DMD patients develop cardiomyopathy, which significantly contributes to mortality. Antisense oligonucleotides (AOs) are a promising DMD therapy, restoring functional dystrophin protein by exon skipping. However, a major limitation with current AOs is the absence of dystrophin correction in heart. Pip peptide-AOs demonstrate high activity in cardiac muscle. To determine their therapeutic value, dystrophic mdx mice were subject to forced exercise to model the DMD cardiac phenotype. Repeated peptide-AO treatments resulted in high levels of cardiac dystrophin protein, which prevented the exercised induced progression of cardiomyopathy, normalising heart size as well as stabilising other cardiac parameters. Treated mice also exhibited significantly reduced cardiac fibrosis and improved sarcolemmal integrity. This work demonstrates that high levels of cardiac dystrophin restored by Pip peptide-AOs prevents further deterioration of cardiomyopathy and pathology following exercise in dystrophic DMD mice.
Collapse
Affiliation(s)
- Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Amer F Saleh
- 1] Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK [2] AstraZeneca R&D, Discovery Safety, Drug safety and Metabolism, Alderley Park, Macclesfield, SK10 4TG, UK
| | - Carolyn A Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Suzan M Hammond
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Caroline Godfrey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Miguel A Varela
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Thomas C Roberts
- 1] Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX [2] Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 NTorrey Pines Road, La Jolla, CA 92037, USA
| | - Kieran Clarke
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| | - Michael J Gait
- Medical Research Council, Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge, CB2 0QH, UK
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK, OX1 3QX
| |
Collapse
|
47
|
Kornegay JN, Spurney CF, Nghiem PP, Brinkmeyer-Langford CL, Hoffman EP, Nagaraju K. Pharmacologic management of Duchenne muscular dystrophy: target identification and preclinical trials. ILAR J 2015; 55:119-49. [PMID: 24936034 DOI: 10.1093/ilar/ilu011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked human disorder in which absence of the protein dystrophin causes degeneration of skeletal and cardiac muscle. For the sake of treatment development, over and above definitive genetic and cell-based therapies, there is considerable interest in drugs that target downstream disease mechanisms. Drug candidates have typically been chosen based on the nature of pathologic lesions and presumed underlying mechanisms and then tested in animal models. Mammalian dystrophinopathies have been characterized in mice (mdx mouse) and dogs (golden retriever muscular dystrophy [GRMD]). Despite promising results in the mdx mouse, some therapies have not shown efficacy in DMD. Although the GRMD model offers a higher hurdle for translation, dogs have primarily been used to test genetic and cellular therapies where there is greater risk. Failed translation of animal studies to DMD raises questions about the propriety of methods and models used to identify drug targets and test efficacy of pharmacologic intervention. The mdx mouse and GRMD dog are genetically homologous to DMD but not necessarily analogous. Subcellular species differences are undoubtedly magnified at the whole-body level in clinical trials. This problem is compounded by disparate cultures in clinical trials and preclinical studies, pointing to a need for greater rigor and transparency in animal experiments. Molecular assays such as mRNA arrays and genome-wide association studies allow identification of genetic drug targets more closely tied to disease pathogenesis. Genes in which polymorphisms have been directly linked to DMD disease progression, as with osteopontin, are particularly attractive targets.
Collapse
|
48
|
Wynne GM, Russell AJ. Drug Discovery Approaches for Rare Neuromuscular Diseases. ORPHAN DRUGS AND RARE DISEASES 2014. [DOI: 10.1039/9781782624202-00257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Rare neuromuscular diseases encompass many diverse and debilitating musculoskeletal disorders, ranging from ultra-orphan conditions that affect only a few families, to the so-called ‘common’ orphan diseases like Duchenne muscular dystrophy (DMD) and spinal muscular atrophy (SMA), which affect several thousand individuals worldwide. Increasingly, pharmaceutical and biotechnology companies, in an effort to improve productivity and rebuild dwindling pipelines, are shifting their business models away from the formerly popular ‘blockbuster’ strategy, with rare diseases being an area of increased focus in recent years. As a consequence of this paradigm shift, coupled with high-profile campaigns by not-for-profit organisations and patient advocacy groups, rare neuromuscular diseases are attracting considerable attention as new therapeutic areas for improved drug therapy. Much pioneering work has taken place to elucidate the underlying pathological mechanisms of many rare neuromuscular diseases. This, in conjunction with the availability of new screening technologies, has inspired the development of several truly innovative therapeutic strategies aimed at correcting the underlying pathology. A survey of medicinal chemistry approaches and the resulting clinical progress for new therapeutic agents targeting this devastating class of degenerative diseases is presented, using DMD and SMA as examples. Complementary strategies using small-molecule drugs and biological agents are included.
Collapse
Affiliation(s)
- Graham M. Wynne
- Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Angela J. Russell
- Chemistry Research Laboratory, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
49
|
Proteomic profiling of the dystrophin-deficient mdx phenocopy of dystrophinopathy-associated cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:246195. [PMID: 24772416 PMCID: PMC3977469 DOI: 10.1155/2014/246195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/16/2014] [Indexed: 01/07/2023]
Abstract
Cardiorespiratory complications are frequent symptoms of Duchenne muscular dystrophy, a neuromuscular disorder caused by primary abnormalities in the dystrophin gene. Loss of cardiac dystrophin initially leads to changes in dystrophin-associated glycoproteins and subsequently triggers secondarily sarcolemmal disintegration, fibre necrosis, fibrosis, fatty tissue replacement, and interstitial inflammation. This results in progressive cardiac disease, which is the cause of death in a considerable number of patients afflicted with X-linked muscular dystrophy. In order to better define the molecular pathogenesis of this type of cardiomyopathy, several studies have applied mass spectrometry-based proteomics to determine proteome-wide alterations in dystrophinopathy-associated cardiomyopathy. Proteomic studies included both gel-based and label-free mass spectrometric surveys of dystrophin-deficient heart muscle from the established mdx animal model of dystrophinopathy. Comparative cardiac proteomics revealed novel changes in proteins associated with mitochondrial energy metabolism, glycolysis, signaling, iron binding, antibody response, fibre contraction, basal lamina stabilisation, and cytoskeletal organisation. This review summarizes the importance of studying cardiomyopathy within the field of muscular dystrophy research, outlines key features of the mdx heart and its suitability as a model system for studying cardiac pathogenesis, and discusses the impact of recent proteomic findings for exploring molecular and cellular aspects of cardiac abnormalities in inherited muscular dystrophies.
Collapse
|
50
|
Oxidative stress in muscular dystrophy: from generic evidence to specific sources and targets. J Muscle Res Cell Motil 2014; 35:23-36. [PMID: 24619215 DOI: 10.1007/s10974-014-9380-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 02/19/2014] [Indexed: 01/06/2023]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases that share a common end-point represented by muscular wasting. MDs are caused by mutations in a variety of genes encoding for different molecules, including extracellular matrix, transmembrane and membrane-associated proteins, cytoplasmic enzymes and nuclear proteins. However, it is still to be elucidated how genetic mutations can affect the molecular mechanisms underlying the contractile impairment occurring in these complex pathologies. The intracellular accumulation of reactive oxygen species (ROS) is widely accepted to play a key role in contractile derangements occurring in the different forms of MDs. However, scarce information is available concerning both the most relevant sources of ROS and their major molecular targets. This review focuses on (i) the sources of ROS, with a special emphasis on monoamine oxidase, a mitochondrial enzyme, and (ii) the targets of ROS, highlighting the relevance of the oxidative modification of myofilament proteins.
Collapse
|