1
|
Liu Y, Li S, Robertson R, Granet JA, Aubry I, Filippelli RL, Tremblay ML, Chang NC. PTPN1/2 inhibition promotes muscle stem cell differentiation in Duchenne muscular dystrophy. Life Sci Alliance 2025; 8:e202402831. [PMID: 39477543 PMCID: PMC11527974 DOI: 10.26508/lsa.202402831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal disease caused by mutations in the DMD gene that encodes dystrophin. Dystrophin deficiency also impacts muscle stem cells (MuSCs), resulting in impaired asymmetric stem cell division and myogenic commitment. Using MuSCs from DMD patients and the DMD mouse model mdx, we found that PTPN1 phosphatase expression is up-regulated and STAT3 phosphorylation is concomitantly down-regulated in DMD MuSCs. To restore STAT3-mediated myogenic signaling, we examined the effect of K884, a novel PTPN1/2 inhibitor, on DMD MuSCs. Treatment with K884 enhanced STAT3 phosphorylation and promoted myogenic differentiation of DMD patient-derived MuSCs. In MuSCs from mdx mice, K884 treatment increased the number of asymmetric cell divisions, correlating with enhanced myogenic differentiation. Interestingly, the pro-myogenic effect of K884 is specific to human and murine DMD MuSCs and is absent from control MuSCs. Moreover, PTPN1/2 loss-of-function experiments indicate that the pro-myogenic impact of K884 is mediated mainly through PTPN1. We propose that PTPN1/2 inhibition may serve as a therapeutic strategy to restore the myogenic function of MuSCs in DMD.
Collapse
MESH Headings
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Animals
- Cell Differentiation/drug effects
- Humans
- Mice
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics
- Mice, Inbred mdx
- STAT3 Transcription Factor/metabolism
- Stem Cells/metabolism
- Stem Cells/cytology
- Muscle Development/genetics
- Muscle Development/drug effects
- Disease Models, Animal
- Phosphorylation
- Signal Transduction/drug effects
- Muscle, Skeletal/metabolism
Collapse
Affiliation(s)
- Yiyang Liu
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Shulei Li
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- https://ror.org/01pxwe438 Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Rebecca Robertson
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Jules A Granet
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Isabelle Aubry
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- https://ror.org/01pxwe438 Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Romina L Filippelli
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Michel L Tremblay
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- https://ror.org/01pxwe438 Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Natasha C Chang
- https://ror.org/01pxwe438 Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
- https://ror.org/01pxwe438 Goodman Cancer Institute, McGill University, Montréal, Canada
| |
Collapse
|
2
|
Kodippili K, Hakim CH, Burke MJ, Yue Y, Teixeira JA, Zhang K, Yao G, Babu GJ, Herzog RW, Duan D. SERCA2a overexpression improves muscle function in a canine Duchenne muscular dystrophy model. Mol Ther Methods Clin Dev 2024; 32:101268. [PMID: 38911286 PMCID: PMC11190715 DOI: 10.1016/j.omtm.2024.101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/16/2024] [Indexed: 06/25/2024]
Abstract
Excessive cytosolic calcium accumulation contributes to muscle degeneration in Duchenne muscular dystrophy (DMD). Sarco/endoplasmic reticulum calcium ATPase (SERCA) is a sarcoplasmic reticulum (SR) calcium pump that actively transports calcium from the cytosol into the SR. We previously showed that adeno-associated virus (AAV)-mediated SERCA2a therapy reduced cytosolic calcium overload and improved muscle and heart function in the murine DMD model. Here, we tested whether AAV SERCA2a therapy could ameliorate muscle disease in the canine DMD model. 7.83 × 1013 vector genome particles of the AAV vector were injected into the extensor carpi ulnaris (ECU) muscles of four juvenile affected dogs. Contralateral ECU muscles received excipient. Three months later, we observed widespread transgene expression and significantly increased SERCA2a levels in the AAV-injected muscles. Treatment improved SR calcium uptake, significantly reduced calpain activity, significantly improved contractile kinetics, and significantly enhanced resistance to eccentric contraction-induced force loss. Nonetheless, muscle histology was not improved. To evaluate the safety of AAV SERCA2a therapy, we delivered the vector to the ECU muscle of adult normal dogs. We achieved strong transgene expression without altering muscle histology and function. Our results suggest that AAV SERCA2a therapy has the potential to improve muscle performance in a dystrophic large mammal.
Collapse
Affiliation(s)
- Kasun Kodippili
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Chady H. Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Matthew J. Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - James A. Teixeira
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Keqing Zhang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
| | - Gang Yao
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
| | - Gopal J. Babu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Roland W. Herzog
- Herman B Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Chemical and Biomedical Engineering, College of Engineering, The University of Missouri, Columbia, MO 65212, USA
- Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO 65212, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
3
|
Jollet M, Mariadassou M, Rué O, Pessemesse L, Ollendorff V, Ramdani S, Vernus B, Bonnieu A, Bertrand-Gaday C, Goustard B, Koechlin-Ramonatxo C. Insight into the Role of Gut Microbiota in Duchenne Muscular Dystrophy: An Age-Related Study in Mdx Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:264-279. [PMID: 37981219 DOI: 10.1016/j.ajpath.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 10/06/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023]
Abstract
Dystrophin deficiency alters the sarcolemma structure, leading to muscle dystrophy, muscle disuse, and ultimately death. Beyond limb muscle deficits, patients with Duchenne muscular dystrophy have numerous transit disorders. Many studies have highlighted the strong relationship between gut microbiota and skeletal muscle. The aims of this study were: i) to characterize the gut microbiota composition over time up to 1 year in dystrophin-deficient mdx mice, and ii) to analyze the intestine structure and function and expression of genes linked to bacterial-derived metabolites in ileum, blood, and skeletal muscles to study interorgan interactions. Mdx mice displayed a significant reduction in the overall number of different operational taxonomic units and their abundance (α-diversity). Mdx genotype predicted 20% of β-diversity divergence, with a large taxonomic modification of Actinobacteria, Proteobacteria, Tenericutes, and Deferribacteres phyla and the included genera. Interestingly, mdx intestinal motility and gene expressions of tight junction and Ffar2 receptor were down-regulated in the ileum. Concomitantly, circulating inflammatory markers related to gut microbiota (tumor necrosis factor, IL-6, monocyte chemoattractant protein-1) and muscle inflammation Tlr4/Myd88 pathway (Toll-like receptor 4, which recognizes pathogen-associated molecular patterns) were up-regulated. Finally, in mdx mice, adiponectin was reduced in blood and its receptor modulated in muscles. This study highlights a specific gut microbiota composition and highlights interorgan interactions in mdx physiopathology with gut microbiota as the potential central metabolic organ.
Collapse
Affiliation(s)
- Maxence Jollet
- DMEM, Université de Montpellier, INRAE, Montpellier, France.
| | - Mahendra Mariadassou
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France; Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | - Olivier Rué
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France; Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | | | | | | | - Barbara Vernus
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | - Anne Bonnieu
- DMEM, Université de Montpellier, INRAE, Montpellier, France
| | | | | | | |
Collapse
|
4
|
Gorza L, Germinario E, Vitadello M, Guerra I, De Majo F, Gasparella F, Caliceti P, Vitiello L, Danieli-Betto D. Curcumin Administration Improves Force of mdx Dystrophic Diaphragm by Acting on Fiber-Type Composition, Myosin Nitrotyrosination and SERCA1 Protein Levels. Antioxidants (Basel) 2023; 12:1181. [PMID: 37371910 DOI: 10.3390/antiox12061181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
The vegetal polyphenol curcumin displays beneficial effects against skeletal muscle derangement induced by oxidative stress, disuse or aging. Since oxidative stress and inflammation are involved in the progression of muscle dystrophy, the effects of curcumin administration were investigated in the diaphragm of mdx mice injected intraperitoneally or subcutaneously with curcumin for 4-12-24 weeks. Curcumin treatment independently of the way and duration of administration (i) ameliorated myofiber maturation index without affecting myofiber necrosis, inflammation and degree of fibrosis; (ii) counteracted the decrease in type 2X and 2B fiber percentage; (iii) increased about 30% both twitch and tetanic tensions of diaphragm strips; (iv) reduced myosin nitrotyrosination and tropomyosin oxidation; (v) acted on two opposite nNOS regulators by decreasing active AMP-Kinase and increasing SERCA1 protein levels, the latter effect being detectable also in myotube cultures from mdx satellite cells. Interestingly, increased contractility, decreased myosin nitrotyrosination and SERCA1 upregulation were also detectable in the mdx diaphragm after a 4-week administration of the NOS inhibitor 7-Nitroindazole, and were not improved further by a combined treatment. In conclusion, curcumin has beneficial effects on the dystrophic muscle, mechanistically acting for the containment of a deregulated nNOS activity.
Collapse
Affiliation(s)
- Luisa Gorza
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Elena Germinario
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Maurizio Vitadello
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Irene Guerra
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Federica De Majo
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | | | - Paolo Caliceti
- Department of Pharmaceutical Sciences, University of Padova, 35131 Padova, Italy
| | - Libero Vitiello
- Department of Biology, University of Padova, 35131 Padova, Italy
| | | |
Collapse
|
5
|
Eisen B, Binah O. Modeling Duchenne Muscular Dystrophy Cardiomyopathy with Patients' Induced Pluripotent Stem-Cell-Derived Cardiomyocytes. Int J Mol Sci 2023; 24:ijms24108657. [PMID: 37240001 DOI: 10.3390/ijms24108657] [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: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked progressive muscle degenerative disease caused by mutations in the dystrophin gene, resulting in death by the end of the third decade of life at the latest. A key aspect of the DMD clinical phenotype is dilated cardiomyopathy, affecting virtually all patients by the end of the second decade of life. Furthermore, despite respiratory complications still being the leading cause of death, with advancements in medical care in recent years, cardiac involvement has become an increasing cause of mortality. Over the years, extensive research has been conducted using different DMD animal models, including the mdx mouse. While these models present certain important similarities to human DMD patients, they also have some differences which pose a challenge to researchers. The development of somatic cell reprograming technology has enabled generation of human induced pluripotent stem cells (hiPSCs) which can be differentiated into different cell types. This technology provides a potentially endless pool of human cells for research. Furthermore, hiPSCs can be generated from patients, thus providing patient-specific cells and enabling research tailored to different mutations. DMD cardiac involvement has been shown in animal models to include changes in gene expression of different proteins, abnormal cellular Ca2+ handling, and other aberrations. To gain a better understanding of the disease mechanisms, it is imperative to validate these findings in human cells. Furthermore, with the recent advancements in gene-editing technology, hiPSCs provide a valuable platform for research and development of new therapies including the possibility of regenerative medicine. In this article, we review the DMD cardiac-related research performed so far using human hiPSCs-derived cardiomyocytes (hiPSC-CMs) carrying DMD mutations.
Collapse
Affiliation(s)
- Binyamin Eisen
- Cardiac Research Laboratory, Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| | - Ofer Binah
- Cardiac Research Laboratory, Department of Physiology, Biophysics and Systems Biology, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
6
|
CRISPR-Based Therapeutic Gene Editing for Duchenne Muscular Dystrophy: Advances, Challenges and Perspectives. Cells 2022; 11:cells11192964. [PMID: 36230926 PMCID: PMC9564082 DOI: 10.3390/cells11192964] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe neuromuscular disease arising from loss-of-function mutations in the dystrophin gene and characterized by progressive muscle degeneration, respiratory insufficiency, cardiac failure, and premature death by the age of thirty. Albeit DMD is one of the most common types of fatal genetic diseases, there is no curative treatment for this devastating disorder. In recent years, gene editing via the clustered regularly interspaced short palindromic repeats (CRISPR) system has paved a new path toward correcting pathological mutations at the genetic source, thus enabling the permanent restoration of dystrophin expression and function throughout the musculature. To date, the therapeutic benefits of CRISPR genome-editing systems have been successfully demonstrated in human cells, rodents, canines, and piglets with diverse DMD mutations. Nevertheless, there remain some nonignorable challenges to be solved before the clinical application of CRISPR-based gene therapy. Herein, we provide an overview of therapeutic CRISPR genome-editing systems, summarize recent advancements in their applications in DMD contexts, and discuss several potential obstacles lying ahead of clinical translation.
Collapse
|
7
|
García-Castañeda M, Michelucci A, Zhao N, Malik S, Dirksen RT. Postdevelopmental knockout of Orai1 improves muscle pathology in a mouse model of Duchenne muscular dystrophy. J Gen Physiol 2022; 154:213383. [PMID: 35939054 PMCID: PMC9365874 DOI: 10.1085/jgp.202213081] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked disorder caused by loss-of-function mutations in the dystrophin gene, is characterized by progressive muscle degeneration and weakness. Enhanced store-operated Ca2+ entry (SOCE), a Ca2+ influx mechanism coordinated by STIM1 sensors of luminal Ca2+ within the sarcoplasmic reticulum (SR) and Ca2+-permeable Orai1 channels in the sarcolemma, is proposed to contribute to Ca2+-mediated muscle damage in DMD. To directly determine the impact of Orai1-dependent SOCE on the dystrophic phenotype, we crossed mdx mice with tamoxifen-inducible, muscle-specific Orai1 knockout mice (mdx-Orai1 KO mice). Both constitutive and SOCE were significantly increased in flexor digitorum brevis fibers from mdx mice, while SOCE was absent in fibers from both Orai1 KO and mdx-Orai1 KO mice. Compared with WT mice, fibers from mdx mice exhibited (1) increased resting myoplasmic Ca2+ levels, (2) reduced total releasable Ca2+ store content, and (3) a prolonged rate of electrically evoked Ca2+ transient decay. These effects were partially normalized in fibers from mdx-Orai1 KO mice. Intact extensor digitorum longus muscles from mdx mice exhibited a significant reduction of maximal specific force, which was rescued in muscles from mdx-Orai1 KO mice. Finally, during exposure to consecutive eccentric contractions, muscles from mdx mice displayed a more pronounced decline in specific force compared with that of WT mice, which was also significantly attenuated by Orai1 ablation. Together, these results indicate that enhanced Orai1-dependent SOCE exacerbates the dystrophic phenotype and that Orai1 deficiency improves muscle pathology by both normalizing Ca2+ homeostasis and promoting sarcolemmal integrity/stability.
Collapse
Affiliation(s)
- Maricela García-Castañeda
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Antonio Michelucci
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY,Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
8
|
Laurila PP, Luan P, Wohlwend M, Zanou N, Crisol B, Imamura de Lima T, Goeminne LJE, Gallart-Ayala H, Shong M, Ivanisevic J, Place N, Auwerx J. Inhibition of sphingolipid de novo synthesis counteracts muscular dystrophy. SCIENCE ADVANCES 2022; 8:eabh4423. [PMID: 35089797 PMCID: PMC8797791 DOI: 10.1126/sciadv.abh4423] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/01/2021] [Indexed: 05/29/2023]
Abstract
Duchenne muscular dystrophy (DMD), the most common muscular dystrophy, is a severe muscle disorder, causing muscle weakness, loss of independence, and premature death. Here, we establish the link between sphingolipids and muscular dystrophy. Transcripts of sphingolipid de novo biosynthesis pathway are up-regulated in skeletal muscle of patients with DMD and other muscular dystrophies, which is accompanied by accumulation of metabolites of the sphingolipid pathway in muscle and plasma. Pharmacological inhibition of sphingolipid synthesis by myriocin in the mdx mouse model of DMD ameliorated the loss in muscle function while reducing inflammation, improving Ca2+ homeostasis, preventing fibrosis of the skeletal muscle, heart, and diaphragm, and restoring the balance between M1 and M2 macrophages. Myriocin alleviated the DMD phenotype more than glucocorticoids. Our study identifies inhibition of sphingolipid synthesis, targeting multiple pathogenetic pathways simultaneously, as a strong candidate for treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ludger J. E. Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
9
|
Meyer P, Notarnicola C, Meli AC, Matecki S, Hugon G, Salvador J, Khalil M, Féasson L, Cances C, Cottalorda J, Desguerre I, Cuisset JM, Sabouraud P, Lacampagne A, Chevassus H, Rivier F, Carnac G. Skeletal Ryanodine Receptors Are Involved in Impaired Myogenic Differentiation in Duchenne Muscular Dystrophy Patients. Int J Mol Sci 2021; 22:12985. [PMID: 34884796 PMCID: PMC8657486 DOI: 10.3390/ijms222312985] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle wasting following repeated muscle damage and inadequate regeneration. Impaired myogenesis and differentiation play a major role in DMD as well as intracellular calcium (Ca2+) mishandling. Ca2+ release from the sarcoplasmic reticulum is mostly mediated by the type 1 ryanodine receptor (RYR1) that is required for skeletal muscle differentiation in animals. The study objective was to determine whether altered RYR1-mediated Ca2+ release contributes to myogenic differentiation impairment in DMD patients. The comparison of primary cultured myoblasts from six boys with DMD and five healthy controls highlighted delayed myoblast differentiation in DMD. Silencing RYR1 expression using specific si-RNA in a healthy control induced a similar delayed differentiation. In DMD myotubes, resting intracellular Ca2+ concentration was increased, but RYR1-mediated Ca2+ release was not changed compared with control myotubes. Incubation with the RYR-calstabin interaction stabilizer S107 decreased resting Ca2+ concentration in DMD myotubes to control values and improved calstabin1 binding to the RYR1 complex. S107 also improved myogenic differentiation in DMD. Furthermore, intracellular Ca2+ concentration was correlated with endomysial fibrosis, which is the only myopathologic parameter associated with poor motor outcome in patients with DMD. This suggested a potential relationship between RYR1 dysfunction and motor impairment. Our study highlights RYR1-mediated Ca2+ leakage in human DMD myotubes and its key role in myogenic differentiation impairment. RYR1 stabilization may be an interesting adjunctive therapeutic strategy in DMD.
Collapse
Affiliation(s)
- Pierre Meyer
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
- Reference Centre for Neuromuscular Diseases AOC, Clinical Investigation Centre, Pediatric Neurology Department, Montpellier University Hospital, 34000 Montpellier, France
| | - Cécile Notarnicola
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Stefan Matecki
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Gérald Hugon
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Jérémy Salvador
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Mirna Khalil
- Clinical Investigation Center, Montpellier University Hospital, 34000 Montpellier, France; (M.K.); (H.C.)
| | - Léonard Féasson
- Myology Unit, Reference Center for Neuromuscular Diseases Euro-NmD, Inter-University Laboratory of Human Movement Sciences—EA7424, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France;
| | - Claude Cances
- Reference Center for Neuromuscular Diseases AOC, Pediatric Neurology Department, Toulouse University Hospital, 3100 Toulouse, France;
- Pediatric Clinical Research Unit, Pediatric Multi-thematic Module CIC 1436, Toulouse Children’s Hospital, 31300 Toulouse, France
| | - Jérôme Cottalorda
- Pediatric Orthopedic and Plastic Surgery Department, Montpellier University Hospital, 34295 Montpellier, France;
| | - Isabelle Desguerre
- Reference Center for Neuromuscular Diseases Paris Nord-Ile-de-France-Est, Pediatric Neurology Department, Necker Enfant Malades University Hospital, Assistance Publique des Hôpitaux de Paris Centre, Paris University, 75019 Paris, France;
| | - Jean-Marie Cuisset
- Reference Center for Neuromuscular Diseases Nord-Ile-de-France-Est, Pediatric Neurology Department, Lille University Hospital, 59000 Lille, France;
| | - Pascal Sabouraud
- Reference Center for Neuromuscular Diseases Nord-Ile-de-France-Est, Pediatric Neurology Department, Reims University Hospital, 51100 Reims, France;
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Hugues Chevassus
- Clinical Investigation Center, Montpellier University Hospital, 34000 Montpellier, France; (M.K.); (H.C.)
| | - François Rivier
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
- Reference Centre for Neuromuscular Diseases AOC, Clinical Investigation Centre, Pediatric Neurology Department, Montpellier University Hospital, 34000 Montpellier, France
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| |
Collapse
|
10
|
Ebrahimi M, Lad H, Fusto A, Tiper Y, Datye A, Nguyen CT, Jacques E, Moyle LA, Nguyen T, Musgrave B, Chávez-Madero C, Bigot A, Chen C, Turner S, Stewart BA, Pegoraro E, Vitiello L, Gilbert PM. De novo revertant fiber formation and therapy testing in a 3D culture model of Duchenne muscular dystrophy skeletal muscle. Acta Biomater 2021; 132:227-244. [PMID: 34048976 DOI: 10.1016/j.actbio.2021.05.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
The biological basis of Duchenne muscular dystrophy (DMD) pathology is only partially characterized and there are still few disease-modifying therapies available, therein underlying the value of strategies to model and study DMD. Dystrophin, the causative gene of DMD, is responsible for linking the cytoskeleton of muscle fibers to the extracellular matrix beyond the sarcolemma. We posited that disease-associated phenotypes not yet captured by two-dimensional culture methods would arise by generating multinucleated muscle cells within a three-dimensional (3D) extracellular matrix environment. Herein we report methods to produce 3D human skeletal muscle microtissues (hMMTs) using clonal, immortalized myoblast lines established from healthy and DMD donors. We also established protocols to evaluate immortalized hMMT self-organization and myotube maturation, as well as calcium handling, force generation, membrane stability (i.e., creatine kinase activity and Evans blue dye permeability) and contractile apparatus organization following electrical-stimulation. In examining hMMTs generated with a cell line wherein the dystrophin gene possessed a duplication of exon 2, we observed rare dystrophin-positive myotubes, which were not seen in 2D cultures. Further, we show that treating DMD hMMTs with a β1-integrin activating antibody, improves contractile apparatus maturation and stability. Hence, immortalized myoblast-derived DMD hMMTs offer a pre-clinical system with which to investigate the potential of duplicated exon skipping strategies and those that protect muscle cells from contraction-induced injury. STATEMENT OF SIGNIFICANCE: Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disorder that is caused by mutation of the dystrophin gene. The biological basis of DMD pathology is only partially characterized and there is no cure for this fatal disease. Here we report a method to produce 3D human skeletal muscle microtissues (hMMTs) using immortalized human DMD and healthy myoblasts. Morphological and functional assessment revealed DMD-associated pathophysiology including impaired calcium handling and de novo formation of dystrophin-positive revertant muscle cells in immortalized DMD hMMTs harbouring an exon 2 duplication, a feature of many DMD patients that has not been recapitulated in culture prior to this report. We further demonstrate that this "DMD in a dish" system can be used as a pre-clinical assay to test a putative DMD therapeutic and study the mechanism of action.
Collapse
Affiliation(s)
- Majid Ebrahimi
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Heta Lad
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Aurora Fusto
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Yekaterina Tiper
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Asiman Datye
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Christine T Nguyen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Erik Jacques
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Thy Nguyen
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Brennen Musgrave
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Carolina Chávez-Madero
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Anne Bigot
- Sorbonne Universite, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris UMRS974, France
| | - Chun Chen
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Scott Turner
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Bryan A Stewart
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Libero Vitiello
- Department of Biology, University of Padua, Padua 35131, Italy; Interuniversity Institute of Myology (IIM), Italy
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada.
| |
Collapse
|
11
|
Al Tanoury Z, Zimmerman JF, Rao J, Sieiro D, McNamara HM, Cherrier T, Rodríguez-delaRosa A, Hick-Colin A, Bousson F, Fugier-Schmucker C, Marchiano F, Habermann B, Chal J, Nesmith AP, Gapon S, Wagner E, Gupta VA, Bassel-Duby R, Olson EN, Cohen AE, Parker KK, Pourquié O. Prednisolone rescues Duchenne muscular dystrophy phenotypes in human pluripotent stem cell-derived skeletal muscle in vitro. Proc Natl Acad Sci U S A 2021; 118:e2022960118. [PMID: 34260377 PMCID: PMC8285911 DOI: 10.1073/pnas.2022960118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating genetic disease leading to degeneration of skeletal muscles and premature death. How dystrophin absence leads to muscle wasting remains unclear. Here, we describe an optimized protocol to differentiate human induced pluripotent stem cells (iPSC) to a late myogenic stage. This allows us to recapitulate classical DMD phenotypes (mislocalization of proteins of the dystrophin-associated glycoprotein complex, increased fusion, myofiber branching, force contraction defects, and calcium hyperactivation) in isogenic DMD-mutant iPSC lines in vitro. Treatment of the myogenic cultures with prednisolone (the standard of care for DMD) can dramatically rescue force contraction, fusion, and branching defects in DMD iPSC lines. This argues that prednisolone acts directly on myofibers, challenging the largely prevalent view that its beneficial effects are caused by antiinflammatory properties. Our work introduces a human in vitro model to study the onset of DMD pathology and test novel therapeutic approaches.
Collapse
Affiliation(s)
- Ziad Al Tanoury
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - John F Zimmerman
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Jyoti Rao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Daniel Sieiro
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Harold M McNamara
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Thomas Cherrier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | | | - Fanny Bousson
- Anagenesis Biotechnologies, 67400 Illkirch Graffenstaden, France
| | | | - Fabio Marchiano
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Bianca Habermann
- Aix-Marseille University, CNRS, Institut de Biologie du Développement de Marseille UMR 7288, The Turing Center for Living Systems, 13009 Marseille, France
| | - Jérome Chal
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Alexander P Nesmith
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Svetlana Gapon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Erica Wagner
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
| | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138
- Department of Physics, Harvard University, Cambridge, MA 02138
| | - Kevin Kit Parker
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Boston, MA 02134
| | - Olivier Pourquié
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR 7104, INSERM U964, Université de Strasbourg, 67411 Illkirch Graffenstaden, France;
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| |
Collapse
|
12
|
Stocco A, Smolina N, Sabatelli P, Šileikytė J, Artusi E, Mouly V, Cohen M, Forte M, Schiavone M, Bernardi P. Treatment with a triazole inhibitor of the mitochondrial permeability transition pore fully corrects the pathology of sapje zebrafish lacking dystrophin. Pharmacol Res 2021; 165:105421. [PMID: 33429034 DOI: 10.1016/j.phrs.2021.105421] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/28/2022]
Abstract
High-throughput screening identified isoxazoles as potent but metabolically unstable inhibitors of the mitochondrial permeability transition pore (PTP). Here we have studied the effects of a metabolically stable triazole analog, TR001, which maintains the PTP inhibitory properties with an in vitro potency in the nanomolar range. We show that TR001 leads to recovery of muscle structure and function of sapje zebrafish, a severe model of Duchenne muscular dystrophy (DMD). PTP inhibition fully restores the otherwise defective respiration in vivo, allowing normal development of sapje individuals in spite of lack of dystrophin. About 80 % sapje zebrafish treated with TR001 are alive and normal at 18 days post fertilization (dpf), a point in time when not a single untreated sapje individual survives. Time to 50 % death of treated zebrafish increases from 5 to 28 dpf, a sizeable number of individuals becoming young adults in spite of the persistent lack of dystrophin expression. TR001 improves respiration of myoblasts and myotubes from DMD patients, suggesting that PTP-dependent dysfunction also occurs in the human disease and that mitochondrial therapy of DMD with PTP-inhibiting triazoles is a viable treatment option.
Collapse
Affiliation(s)
- Anna Stocco
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Padova, Italy
| | - Natalia Smolina
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Padova, Italy
| | - Patrizia Sabatelli
- CNR-Institute of Molecular Genetics "Luigi Luca Cavalli-Sforza"-Unit of Bologna, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Justina Šileikytė
- Vollum Institute and Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Edoardo Artusi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Padova, Italy
| | - Vincent Mouly
- Center for Research in Myology UMRS 974, Sorbonne Université, INSERM, Myology Institute, Paris, France
| | - Michael Cohen
- Vollum Institute and Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Michael Forte
- Vollum Institute and Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, USA
| | - Marco Schiavone
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Padova, Italy.
| | - Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Padova, Italy.
| |
Collapse
|
13
|
Balderas-Villalobos J, Steele TWE, Eltit JM. Physiological and Pathological Relevance of Selective and Nonselective Ca 2+ Channels in Skeletal and Cardiac Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:225-247. [PMID: 35138617 PMCID: PMC10683374 DOI: 10.1007/978-981-16-4254-8_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Contraction of the striated muscle is fundamental for human existence. The action of voluntary skeletal muscle enables activities such as breathing, establishing body posture, and diverse body movements. Additionally, highly precise motion empowers communication, artistic expression, and other activities that define everyday human life. The involuntary contraction of striated muscle is the core function of the heart and is essential for blood flow. Several ion channels are important in the transduction of action potentials to cytosolic Ca2+ signals that enable muscle contraction; however, other ion channels are involved in the progression of muscle pathologies that can impair normal life or threaten it. This chapter describes types of selective and nonselective Ca2+ permeable ion channels expressed in the striated muscle, their participation in different aspects of muscle excitation and contraction, and their relevance to the progression of some pathological states.
Collapse
Affiliation(s)
- Jaime Balderas-Villalobos
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Tyler W E Steele
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Jose M Eltit
- Department of Physiology and Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
14
|
Nassoro DD, Torres L, Marando R, Mboma L, Mushi S, Habakkuk Mwakyula I. A child with duchenne muscular dystrophy: A case report of a rare diagnosis among Africans. Clin Case Rep 2020; 8:2654-2660. [PMID: 33363799 PMCID: PMC7752564 DOI: 10.1002/ccr3.3254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 01/14/2023] Open
Abstract
In Africa, lack of awareness and low index of suspicion of rare diseases like dystrophinopathies, directly or indirectly, contributes to the increased morbidity and mortality. Therefore, even though the data on prevalence is limited, we need to have a high degree of suspicion in patients presenting with suggestive clinical features.
Collapse
Affiliation(s)
- David D. Nassoro
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Liset Torres
- Department of PathologyMbeya Zonal Referral HospitalMbeyaTanzania
- Department of PathologyThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Rehema Marando
- Department of Pediatrics and Child HealthMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Pediatrics and Child HealthThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Lazaro Mboma
- Department of SurgeryMbeya Zonal Referral HospitalMbeyaTanzania
- Department of SurgeryThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Seraphine Mushi
- Department of PhysiotherapyMbeya Zonal Referral HospitalMbeyaTanzania
| | - Issakwisa Habakkuk Mwakyula
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| |
Collapse
|
15
|
Sztretye M, Szabó L, Dobrosi N, Fodor J, Szentesi P, Almássy J, Magyar ZÉ, Dienes B, Csernoch L. From Mice to Humans: An Overview of the Potentials and Limitations of Current Transgenic Mouse Models of Major Muscular Dystrophies and Congenital Myopathies. Int J Mol Sci 2020; 21:ijms21238935. [PMID: 33255644 PMCID: PMC7728138 DOI: 10.3390/ijms21238935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Muscular dystrophies are a group of more than 160 different human neuromuscular disorders characterized by a progressive deterioration of muscle mass and strength. The causes, symptoms, age of onset, severity, and progression vary depending on the exact time point of diagnosis and the entity. Congenital myopathies are rare muscle diseases mostly present at birth that result from genetic defects. There are no known cures for congenital myopathies; however, recent advances in gene therapy are promising tools in providing treatment. This review gives an overview of the mouse models used to investigate the most common muscular dystrophies and congenital myopathies with emphasis on their potentials and limitations in respect to human applications.
Collapse
|
16
|
Soo TCC, See SA, Bhassu S. Potential muscle activity disturbance in Penaeus monodon during Acute Hepatopancreatic Necrosis Disease (AHPND) infection: Inference through gene expression, calcium concentration, and MicroRNA. J Invertebr Pathol 2020; 177:107497. [PMID: 33130047 DOI: 10.1016/j.jip.2020.107497] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/21/2023]
Abstract
Global shrimp aquaculture farmers have suffered major economic losses due to disease outbreaks. A notable shrimp disease is Acute Hepatopancreatic Necrosis Disease (AHPND), which is caused by a new strain of Vibrio parahaemolyticus bacteria (VpAHPND) that mainly inhabits the shrimp gut and damages the hepatopancreas. Fewer studies have investigated whether this disease will affect shrimp muscle functioning or cause any muscle damage. We challenged Penaeus monodon shrimp with VpAHPND bacteria using an immersion method. Expression of Dystrophin gene, an important regulatory gene for maintenance of muscle integrity, was quantified from muscle samples using qRT-PCR. Additional verification was conducted by determining calcium concentration and bta-miR-4286 and dre-miR-107b miRNAs expression. P. monodon dystrophin gene demonstrated the highest expression level during AHPND infection when muscle calcium concentration was detected at its lowest level at 6 h post-infection (hpi). The highest muscle calcium concentration, determined at 36 hpi, was supported by higher bta-miR-4286 miRNA expression and lower dre-miR-107b miRNA expression in VpAHPND-infected samples compared to uninfected samples at the same time point. We deduced an interactive relationship between dystrophin gene expression, calcium concentration, and miRNA expression in P. monodon muscle tissues triggered by the invading VpAHPND bacterium.
Collapse
Affiliation(s)
- Tze Chiew Christie Soo
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - SiouNing Aileen See
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Subha Bhassu
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
17
|
Ghaleh B, Barthélemy I, Wojcik J, Sambin L, Bizé A, Hittinger L, Tran TD, Thomé FP, Blot S, Su JB. Protective effects of rimeporide on left ventricular function in golden retriever muscular dystrophy dogs. Int J Cardiol 2020; 312:89-95. [PMID: 32199683 DOI: 10.1016/j.ijcard.2020.03.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/24/2020] [Accepted: 03/11/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Alterations in intracellular Na+ and Ca2+ have been observed in patients with Duchenne muscular dystrophy (DMD) and in animal models of DMD, and inhibition of Na+-H+ exchanger 1 (NHE1) by rimeporide has previously demonstrated cardioprotective effects in animal models of myocardial ischemia and heart failure. Since heart failure is becoming a predominant cause of death in DMD patients, this study aimed to demonstrate a cardioprotective effect of chronic administration of rimeporide in a canine model of DMD. METHODS Golden retriever muscular dystrophy (GRMD) dogs were randomized to orally receive rimeporide (10 mg/kg, twice a day) or placebo from 2 months to 1 year of age. Left ventricular (LV) function was assessed by conventional and advanced echocardiography. RESULTS Compared with placebo-treated GRMD, LV function deterioration with age was limited in rimeporide-treated GRMD dogs as indicated by the preservation of LV ejection fraction as well as overall cardiac parameters different from placebo-treated dogs, as revealed by composite cardiac scores and principal component analysis. In addition, principal component analysis clustered rimeporide-treated GRMD dogs close to healthy control dogs. CONCLUSIONS Chronic administration of the NHE1 inhibitor rimeporide exerted a protective effect against LV function decline in GRMD dogs. This study provides proof of concept to explore the cardiac effects of rimeporide in DMD patients.
Collapse
Affiliation(s)
- Bijan Ghaleh
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Inès Barthélemy
- U955-IMRB, Equipe 10, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jérôme Wojcik
- Translational Informatics & Biometrics Europe, Precision for Medicine, CH-1202 Geneva, Switzerland
| | - Lucien Sambin
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Alain Bizé
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Luc Hittinger
- U955-IMRB, Equipe 10, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France; AP-HP, Hôpitaux Universitaires Henri Mondor, Fédération de Cardiologie, F-94000 Créteil, France
| | - Thien Duc Tran
- EspeRare, Campus Biotech Innovation Park, Avenue de Secheron 15, 1202 Geneva, Switzerland
| | - Florence Porte Thomé
- EspeRare, Campus Biotech Innovation Park, Avenue de Secheron 15, 1202 Geneva, Switzerland
| | - Stéphane Blot
- U955-IMRB, Equipe 10, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | - Jin Bo Su
- U955-IMRB, Equipe 03, Inserm, UPEC, Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France.
| |
Collapse
|
18
|
Choi JH, Jeong SY, Oh MR, Allen PD, Lee EH. TRPCs: Influential Mediators in Skeletal Muscle. Cells 2020; 9:cells9040850. [PMID: 32244622 PMCID: PMC7226745 DOI: 10.3390/cells9040850] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
Ca2+ itself or Ca2+-dependent signaling pathways play fundamental roles in various cellular processes from cell growth to death. The most representative example can be found in skeletal muscle cells where a well-timed and adequate supply of Ca2+ is required for coordinated Ca2+-dependent skeletal muscle functions, such as the interactions of contractile proteins during contraction. Intracellular Ca2+ movements between the cytosol and sarcoplasmic reticulum (SR) are strictly regulated to maintain the appropriate Ca2+ supply in skeletal muscle cells. Added to intracellular Ca2+ movements, the contribution of extracellular Ca2+ entry to skeletal muscle functions and its significance have been continuously studied since the early 1990s. Here, studies on the roles of channel proteins that mediate extracellular Ca2+ entry into skeletal muscle cells using skeletal myoblasts, myotubes, fibers, tissue, or skeletal muscle-originated cell lines are reviewed with special attention to the proposed functions of transient receptor potential canonical proteins (TRPCs) as store-operated Ca2+ entry (SOCE) channels under normal conditions and the potential abnormal properties of TRPCs in muscle diseases such as Duchenne muscular dystrophy (DMD).
Collapse
Affiliation(s)
- Jun Hee Choi
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Yeon Jeong
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Mi Ri Oh
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Paul D. Allen
- Leeds Institute of Biomedical & Clinical Sciences, St. James’s University Hospital, University of Leeds, Leeds LS97TF, UK
| | - Eun Hui Lee
- Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-7279
| |
Collapse
|
19
|
Bella P, Farini A, Banfi S, Parolini D, Tonna N, Meregalli M, Belicchi M, Erratico S, D'Ursi P, Bianco F, Legato M, Ruocco C, Sitzia C, Sangiorgi S, Villa C, D'Antona G, Milanesi L, Nisoli E, Mauri P, Torrente Y. Blockade of IGF2R improves muscle regeneration and ameliorates Duchenne muscular dystrophy. EMBO Mol Med 2020; 12:e11019. [PMID: 31793167 PMCID: PMC6949491 DOI: 10.15252/emmm.201911019] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a debilitating fatal X-linked muscle disorder. Recent findings indicate that IGFs play a central role in skeletal muscle regeneration and development. Among IGFs, insulinlike growth factor 2 (IGF2) is a key regulator of cell growth, survival, migration and differentiation. The type 2 IGF receptor (IGF2R) modulates circulating and tissue levels of IGF2 by targeting it to lysosomes for degradation. We found that IGF2R and the store-operated Ca2+ channel CD20 share a common hydrophobic binding motif that stabilizes their association. Silencing CD20 decreased myoblast differentiation, whereas blockade of IGF2R increased proliferation and differentiation in myoblasts via the calmodulin/calcineurin/NFAT pathway. Remarkably, anti-IGF2R induced CD20 phosphorylation, leading to the activation of sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase (SERCA) and removal of intracellular Ca2+ . Interestingly, we found that IGF2R expression was increased in dystrophic skeletal muscle of human DMD patients and mdx mice. Blockade of IGF2R by neutralizing antibodies stimulated muscle regeneration, induced force recovery and normalized capillary architecture in dystrophic mdx mice representing an encouraging starting point for the development of new biological therapies for DMD.
Collapse
Affiliation(s)
- Pamela Bella
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Andrea Farini
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Stefania Banfi
- Hematology Department Fondazione IRCCSDepartment of Oncology and Hemato‐oncologyIstituto Nazionale dei TumoriUniversitá degli Studi di MilanoMilanItaly
| | | | | | - Mirella Meregalli
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Marzia Belicchi
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | | | - Pasqualina D'Ursi
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | | | - Mariella Legato
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Chiara Ruocco
- Department of Medical Biotechnology and Translational MedicineCenter for Study and Research on ObesityMilan UniversityMilanItaly
| | - Clementina Sitzia
- UOC SMEL‐1Scuola di Specializzazione di Patologia Clinica e Biochimica ClinicaUniversità degli Studi di MilanoMilanItaly
| | - Simone Sangiorgi
- Neurosurgery UnitDepartment of SurgeryASST Lariana‐S. Anna HospitalComoItaly
| | - Chiara Villa
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| | - Giuseppe D'Antona
- Department of Public Health, Experimental and Forensic MedicinePavia UniversityPaviaItaly
| | - Luciano Milanesi
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | - Enzo Nisoli
- Department of Medical Biotechnology and Translational MedicineCenter for Study and Research on ObesityMilan UniversityMilanItaly
| | - PierLuigi Mauri
- Institute of Technologies in BiomedicineNational Research Council (ITB‐CNR)MilanItaly
| | - Yvan Torrente
- Stem Cell LaboratoryDepartment of Pathophysiology and TransplantationUnit of NeurologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoCentro Dino FerrariUniversitá degli Studi di MilanoMilanItaly
| |
Collapse
|
20
|
Matre PR, Mu X, Wu J, Danila D, Hall MA, Kolonin MG, Darabi R, Huard J. CRISPR/Cas9-Based Dystrophin Restoration Reveals a Novel Role for Dystrophin in Bioenergetics and Stress Resistance of Muscle Progenitors. Stem Cells 2019; 37:1615-1628. [PMID: 31574188 PMCID: PMC6916636 DOI: 10.1002/stem.3094] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 06/03/2019] [Accepted: 06/26/2019] [Indexed: 12/11/2022]
Abstract
Although the lack of dystrophin expression in muscle myofibers is the central cause of Duchenne muscular dystrophy (DMD), accumulating evidence suggests that DMD may also be a stem cell disease. Recent studies have revealed dystrophin expression in satellite cells and demonstrated that dystrophin deficiency is directly related to abnormalities in satellite cell polarity, asymmetric division, and epigenetic regulation, thus contributing to the manifestation of the DMD phenotype. Although metabolic and mitochondrial dysfunctions have also been associated with the DMD pathophysiology profile, interestingly, the role of dystrophin with respect to stem cells dysfunction has not been elucidated. In the past few years, editing of the gene that encodes dystrophin has emerged as a promising therapeutic approach for DMD, although the effects of dystrophin restoration in stem cells have not been addressed. Herein, we describe our use of a clustered regularly interspaced short palindromic repeats/Cas9‐based system to correct the dystrophin mutation in dystrophic (mdx) muscle progenitor cells (MPCs) and show that the expression of dystrophin significantly improved cellular properties of the mdx MPCs in vitro. Our findings reveal that dystrophin‐restored mdx MPCs demonstrated improvements in cell proliferation, differentiation, bioenergetics, and resistance to oxidative and endoplasmic reticulum stress. Furthermore, our in vivo studies demonstrated improved transplantation efficiency of the corrected MPCs in the muscles of mdx mice. Our results indicate that changes in cellular energetics and stress resistance via dystrophin restoration enhance muscle progenitor cell function, further validating that dystrophin plays a role in stem cell function and demonstrating the potential for new therapeutic approaches for DMD. stem cells2019;37:1615–1628
Collapse
Affiliation(s)
- Polina R Matre
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xiaodong Mu
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA
| | - Jianbo Wu
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Delia Danila
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mary A Hall
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Mikhail G Kolonin
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Radbod Darabi
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Center for Regenerative Sports Medicine, Steadman Philippon Research Institute, Vail, Colorado, USA.,Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
21
|
Role of STIM1/ORAI1-mediated store-operated Ca 2+ entry in skeletal muscle physiology and disease. Cell Calcium 2018; 76:101-115. [PMID: 30414508 DOI: 10.1016/j.ceca.2018.10.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 11/23/2022]
Abstract
Store-operated Ca2+ entry (SOCE) is a Ca2+ entry mechanism activated by depletion of intracellular Ca2+ stores. In skeletal muscle, SOCE is mediated by an interaction between stromal-interacting molecule-1 (STIM1), the Ca2+ sensor of the sarcoplasmic reticulum, and ORAI1, the Ca2+-release-activated-Ca2+ (CRAC) channel located in the transverse tubule membrane. This review focuses on the molecular mechanisms and physiological role of SOCE in skeletal muscle, as well as how alterations in STIM1/ORAI1-mediated SOCE contribute to muscle disease. Recent evidence indicates that SOCE plays an important role in both muscle development/growth and fatigue. The importance of SOCE in muscle is further underscored by the discovery that loss- and gain-of-function mutations in STIM1 and ORAI1 result in an eclectic array of disorders with clinical myopathy as central defining component. Despite differences in clinical phenotype, all STIM1/ORAI1 gain-of-function mutations-linked myopathies are characterized by the abnormal accumulation of intracellular membranes, known as tubular aggregates. Finally, dysfunctional STIM1/ORAI1-mediated SOCE also contributes to the pathogenesis of muscular dystrophy, malignant hyperthermia, and sarcopenia. The picture to emerge is that tight regulation of STIM1/ORAI1-dependent Ca2+ signaling is critical for optimal skeletal muscle development/function such that either aberrant increases or decreases in SOCE activity result in muscle dysfunction.
Collapse
|
22
|
Personalized gene and cell therapy for Duchenne Muscular Dystrophy. Neuromuscul Disord 2018; 28:803-824. [DOI: 10.1016/j.nmd.2018.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 01/09/2023]
|
23
|
Yu HK, Xia B, Liu X, Han C, Chen W, Li Z. Initial application of three-dimensional speckle-tracking echocardiography to detect subclinical left ventricular dysfunction and stratify cardiomyopathy associated with Duchenne muscular dystrophy in children. Int J Cardiovasc Imaging 2018; 35:67-76. [PMID: 30105418 DOI: 10.1007/s10554-018-1436-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 08/04/2018] [Indexed: 01/16/2023]
Abstract
Three-dimensional (3D) speckle-tracking echocardiography (STE) is a new imaging modality used for quantitative analysis of left ventricular (LV) function. The aim of this study is to assess the value of 3D STE in early detection of subclinical myocardial involvement in children with Duchenne muscular dystrophy (DMD). Fifty-six children with DMD (mean age, 8.8 ± 1.9 years) and 31 age-matched control subjects were studied. Patients were subdivided into two groups by age: ≤ 8 or > 8 years. Standard echocardiography examinations were performed to measure LV size and ejection fraction (EF). 3D STE was performed to assess LV 3D global strain and LV end-diastolic volume (EDV), end-systolic volume (ESV), and EF. Standard and 3D echocardiography measures were compared between children with DMD and those in the control group as well as between different patient groups. The areas under the receiver-operating characteristic (ROC) curve were calculated to determine the capability of 3D global strain indices to discriminate between patients and control subjects. No significant difference was detected in either LVEF derived from M-mode or 3D echocardiography between the two groups, and they were both within the normal range. Compared with control subjects, children with DMD had significantly reduced LV 3D global longitudinal strain (GLS; - 16.6 ± 4.7 vs. - 19.5 ± 3.7, p = 0.003), global circumferential strain (GCS; - 13.7 ± 2.9 vs. - 15.8 ± 2.6, p = 0.001), global radial strain (GRS; 42.5 ± 9.7 vs. 50.3 ± 10.4, p = 0.001), and global area strain (GAS; - 25.3 ± 4.9 vs. - 30.7 ± 4.1, p = 0.000). The older DMD children (age > 8 years) had lower GLS (- 15.1 ± 4.43 vs. - 18.6 ± 4.35, p < 0.05), GCS (- 12.8 ± 3.48 vs. - 14.8 ± 2.83, p < 0.001), GAS (- 23.8 ± 4.7 vs. - 29.0 ± 5.4, p < 0.001), and GRS (40.7 ± 8.8 vs. 47.3 ± 11.5, p < 0.05) than younger patients (age ≤ 8 years). The AUC of GAS was 0.80, and the cutoff value of - 29.5 had a sensitivity of 85.7% and a specificity of 71.0% for differentiating DMD patients from control. 3D speckle-tracking echocardiography is useful for detecting subclinical myocardial dysfunction and stratifying cardiomyopathy in children with DMD.
Collapse
Affiliation(s)
- Hong-Kui Yu
- Department of Ultrasonography, Shenzhen Children's Hospital, 7019, Yitian Road, Shenzhen, Guangdong, China
| | - Bei Xia
- Department of Ultrasonography, Shenzhen Children's Hospital, 7019, Yitian Road, Shenzhen, Guangdong, China.
| | - Xiao Liu
- Department of Ultrasonography, Shenzhen Children's Hospital, 7019, Yitian Road, Shenzhen, Guangdong, China
| | - Chunxi Han
- Laboratory of Neuromuscular Disease, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Weiling Chen
- Department of Ultrasonography, Shenzhen Children's Hospital, 7019, Yitian Road, Shenzhen, Guangdong, China
| | - Zhihui Li
- Department of Ultrasonography, Shenzhen Children's Hospital, 7019, Yitian Road, Shenzhen, Guangdong, China
| |
Collapse
|
24
|
Crowe KE, Shao G, Flanigan KM, Martin PT. N-terminal α Dystroglycan (αDG-N): A Potential Serum Biomarker for Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 3:247-260. [PMID: 27854211 DOI: 10.3233/jnd-150127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a severe, progressive, neuromuscular disorder of childhood. While a number of serum factors have been identified as potential biomarkers of DMD, none, as yet, are proteins within the dystrophin-associated glycoprotein (DAG) complex. OBJECTIVE We have developed an immobilized serum ELISA assay to measure the expression of a constitutively cleaved and secreted component of the DAG complex, the N-terminal domain of α dystroglycan (αDG-N), and assayed relative expression in serum from muscular dystrophy patients and normal controls. METHODS ELISAs of immobilized patient or mouse serum and Western blots were used to assess αDG-N expression. RESULTS Immobilization of diluted serum on ELISA plates was important for this assay, as methods to measure serum αDG-N in solution were less robust. αDG-N ELISA signals were significantly reduced in DMD serum (27±3% decrease, n = 9, p < 0.001) relative to serum from otherwise normal controls (n = 38), and calculated serum αDG-N concentrations were reduced in DMD relative to normal (p < 0.01) and Becker Muscular Dystrophy (n = 11, p < 0.05) patient serum. By contrast, ELISA signals from patients with Inclusion Body Myositis were not different than normal (4±3% decrease, n = 8, p = 0.99). αDG-N serum signals were also significantly reduced in utrophin-deficient mdx mice as compared to mdx and wild type mice. CONCLUSIONS Our results are the first demonstration of a component of the DAG complex as a potential serum biomarker in DMD. Such a serum measure could be further developed as a tool to help reflect overall muscle DAG complex expression or stability.
Collapse
Affiliation(s)
- Kelly E Crowe
- Graduate Program in Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Guohong Shao
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
25
|
Iwata Y, Katayama Y, Okuno Y, Wakabayashi S. Novel inhibitor candidates of TRPV2 prevent damage of dystrophic myocytes and ameliorate against dilated cardiomyopathy in a hamster model. Oncotarget 2018; 9:14042-14057. [PMID: 29581825 PMCID: PMC5865651 DOI: 10.18632/oncotarget.24449] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/01/2018] [Indexed: 11/25/2022] Open
Abstract
Transient receptor potential cation channel, subfamily V, member 2 (TRPV2) is a principal candidate for abnormal Ca2+-entry pathways, which is a potential target for therapy of muscular dystrophy and cardiomyopathy. Here, an in silico drug screening and the following cell-based screening to measure the TRPV2 activation were carried out in HEK293 cells expressing TRPV2 using lead compounds (tranilast or SKF96365) and off-patent drug stocks. We identified 4 chemical compounds containing amino-benzoyl groups and 1 compound (lumin) containing an ethylquinolinium group as candidate TRPV2 inhibitors. Three of these compounds inhibited Ca2+ entry through both mouse and human TRPV2, with IC50 of less than 10 μM, but had no apparent effect on other members of TRP family such as TRPV1 and TRPC1. Particularly, lumin inhibited agonist-induced TRPV2 channel activity at a low dose. These compounds inhibited abnormally increased Ca2+ influx and prevented stretch-induced skeletal muscle damage in cultured myocytes from dystrophic hamsters (J2N-k). Further, they ameliorated cardiac dysfunction, and prevented disease progression in vivo in the same J2N-k hamsters developing dilated cardiomyopathy as well as muscular dystrophy. The identified compounds described here are available as experimental tools and represent potential treatments for patients with cardiomyopathy and muscular dystrophy.
Collapse
Affiliation(s)
- Yuko Iwata
- Departments of Molecular Physiology and Clinical Research, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Yoshimi Katayama
- Pharmacological Research Laboratories, Drug Safety Testing Center Co., Ltd., Higashimatsuyama, Saitama, Japan.,Present affiliation: Biological Research Laboratories, Nissan Chemical Industries, Ltd, Shiraoka, Saitama, Japan
| | - Yasushi Okuno
- Department of Clinical System Onco-Informatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigeo Wakabayashi
- Departments of Molecular Physiology and Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.,Present affiliation: Department of Pharmocology, Osaka Medical Collage, Takatsuki, Osaka, Japan
| |
Collapse
|
26
|
Capogrosso RF, Mantuano P, Uaesoontrachoon K, Cozzoli A, Giustino A, Dow T, Srinivassane S, Filipovic M, Bell C, Vandermeulen J, Massari AM, De Bellis M, Conte E, Pierno S, Camerino GM, Liantonio A, Nagaraju K, De Luca A. Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy. FASEB J 2018; 32:1025-1043. [PMID: 29097503 PMCID: PMC5888399 DOI: 10.1096/fj.201700182rrr] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022]
Abstract
Muscle fibers lacking dystrophin undergo a long-term alteration of Ca2+ homeostasis, partially caused by a leaky Ca2+ release ryanodine (RyR) channel. S48168/ARM210, an RyR calcium release channel stabilizer (a Rycal compound), is expected to enhance the rebinding of calstabin to the RyR channel complex and possibly alleviate the pathologic Ca2+ leakage in dystrophin-deficient skeletal and cardiac muscle. This study systematically investigated the effect of S48168/ARM210 on the phenotype of mdx mice by means of a first proof-of-concept, short (4 wk), phase 1 treatment, followed by a 12-wk treatment (phase 2) performed in parallel by 2 independent laboratories. The mdx mice were treated with S48168/ARM210 at two different concentrations (50 or 10 mg/kg/d) in their drinking water for 4 and 12 wk, respectively. The mice were subjected to treadmill sessions twice per week (12 m/min for 30 min) to unmask the mild disease. This testing was followed by in vivo forelimb and hindlimb grip strength and fatigability measurement, ex vivo extensor digitorum longus (EDL) and diaphragm (DIA) force contraction measurement and histologic and biochemical analysis. The treatments resulted in functional (grip strength, ex vivo force production in DIA and EDL muscles) as well as histologic improvement after 4 and 12 wk, with no adverse effects. Furthermore, levels of cellular biomarkers of calcium homeostasis increased. Therefore, these data suggest that S48168/ARM210 may be a safe therapeutic option, at the dose levels tested, for the treatment of Duchenne muscular dystrophy (DMD).-Capogrosso, R. F., Mantuano, P., Uaesoontrachoon, K., Cozzoli, A., Giustino, A., Dow, T., Srinivassane, S., Filipovic, M., Bell, C., Vandermeulen, J., Massari, A. M., De Bellis, M., Conte, E., Pierno, S., Camerino, G. M., Liantonio, A., Nagaraju, K., De Luca, A. Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy.
Collapse
Affiliation(s)
| | - Paola Mantuano
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | | | - Anna Cozzoli
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Arcangela Giustino
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Todd Dow
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | | | - Marina Filipovic
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | - Christina Bell
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | | | - Ada Maria Massari
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Michela De Bellis
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Elena Conte
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Sabata Pierno
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Giulia Maria Camerino
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Antonella Liantonio
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Kanneboyina Nagaraju
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, New York, USA
| | - Annamaria De Luca
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| |
Collapse
|
27
|
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
|
28
|
Tsuda T, Fitzgerald KK. Dystrophic Cardiomyopathy: Complex Pathobiological Processes to Generate Clinical Phenotype. J Cardiovasc Dev Dis 2017; 4:jcdd4030014. [PMID: 29367543 PMCID: PMC5715712 DOI: 10.3390/jcdd4030014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/27/2017] [Accepted: 08/30/2017] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and X-linked dilated cardiomyopathy (XL-DCM) consist of a unique clinical entity, the dystrophinopathies, which are due to variable mutations in the dystrophin gene. Dilated cardiomyopathy (DCM) is a common complication of dystrophinopathies, but the onset, progression, and severity of heart disease differ among these subgroups. Extensive molecular genetic studies have been conducted to assess genotype-phenotype correlation in DMD, BMD, and XL-DCM to understand the underlying mechanisms of these diseases, but the results are not always conclusive, suggesting the involvement of complex multi-layers of pathological processes that generate the final clinical phenotype. Dystrophin protein is a part of dystrophin-glycoprotein complex (DGC) that is localized in skeletal muscles, myocardium, smooth muscles, and neuronal tissues. Diversity of cardiac phenotype in dystrophinopathies suggests multiple layers of pathogenetic mechanisms in forming dystrophic cardiomyopathy. In this review article, we review the complex molecular interactions involving the pathogenesis of dystrophic cardiomyopathy, including primary gene mutations and loss of structural integrity, secondary cellular responses, and certain epigenetic and other factors that modulate gene expressions. Involvement of epigenetic gene regulation appears to lead to specific cardiac phenotypes in dystrophic hearts.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| | - Kristi K Fitzgerald
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, Wilmington, 1600 Rockland Rd, DE 19803, USA.
| |
Collapse
|
29
|
Pauly M, Angebault-Prouteau C, Dridi H, Notarnicola C, Scheuermann V, Lacampagne A, Matecki S, Fauconnier J. ER stress disturbs SR/ER-mitochondria Ca 2+ transfer: Implications in Duchenne muscular dystrophy. Biochim Biophys Acta Mol Basis Dis 2017. [PMID: 28625916 DOI: 10.1016/j.bbadis.2017.06.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Besides its role in calcium (Ca2+) homeostasis, the sarco-endoplamic reticulum (SR/ER) controls protein folding and is tethered to mitochondria. Under pathophysiological conditions the unfolded protein response (UPR) is associated with disturbance in SR/ER-mitochondria crosstalk. Here, we investigated whether ER stress altered SR/ER-mitochondria links, Ca2+ handling and muscle damage in WT (Wild Type) and mdx mice, the murine model of Duchenne Muscular Dystrophy (DMD). In WT mice, the SR/ER-mitochondria links were decreased in isolated FDB muscle fibers after injection of ER stress activator tunicamycin (TM). Ca2+ imaging revealed an increase of cytosolic Ca2+ transient and a decrease of mitochondrial Ca2+ uptake. The force generating capacity of muscle dropped after TM. This impaired contractile function was accompanied by an increase in autophagy markers and calpain-1 activation. Conversely, ER stress inhibitors restored SR/ER-mitochondria links, mitochondrial Ca2+ uptake and improved diaphragm contractility in mdx mice. Our findings demonstrated that ER stress-altered SR/ER-mitochondria links, disturbed Ca2+ handling and muscle function in WT and mdx mice. Thus, ER stress may open up a prospect of new therapeutic targets in DMD.
Collapse
Affiliation(s)
- Marion Pauly
- Inserm U1055, Hypoxie et Physiopathologies, Université Grenoble Alpes, Grenoble, France; Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | | | - Haikel Dridi
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | - Cécile Notarnicola
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | - Valérie Scheuermann
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | - Alain Lacampagne
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | - Stefan Matecki
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France
| | - Jérémy Fauconnier
- Inserm U1046, UMR CNRS 9214, Université Montpellier, CHRU Montpellier, Montpellier, France.
| |
Collapse
|
30
|
Denny AP, Heather AK. Are Antioxidants a Potential Therapy for FSHD? A Review of the Literature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7020295. [PMID: 28690764 PMCID: PMC5485364 DOI: 10.1155/2017/7020295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/27/2017] [Accepted: 05/03/2017] [Indexed: 11/21/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an inherited myopathy affecting approximately 1 in 7500 individuals worldwide. It is a progressive disease characterised by skeletal muscle weakness and wasting. A genetic mutation on the 4q35 chromosome results in the expression of the double homeobox 4 gene (DUX4) which drives oxidative stress, inflammation, toxicity, and atrophy within the skeletal muscle. FSHD is characterised by oxidative stress, and there is currently no cure and a lack of therapies for the disease. Antioxidants have been researched for many years, with investigators aiming to use antioxidants therapeutically for oxidative stress-associated diseases. This has included both natural and synthetic antioxidants. The use of antioxidants in preclinical or clinical models has been largely successful with a plethora of research reporting positive results. However, when translated to clinical trials, the use of antioxidants as a therapeutic intervention for a variety of disease has been largely unsuccessful. Moreover, specifically focusing on FSHD, limited research has been conducted on the use of antioxidants as a therapy in either preclinical or clinical models. This review summarises the current state of antioxidant use in the treatment of FSHD and discusses their potential avenue for therapeutic use for FSHD patients.
Collapse
Affiliation(s)
- Adam Philip Denny
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Alison Kay Heather
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
31
|
Mignen O, Constantin B, Potier-Cartereau M, Penna A, Gautier M, Guéguinou M, Renaudineau Y, Shoji KF, Félix R, Bayet E, Buscaglia P, Debant M, Chantôme A, Vandier C. Constitutive calcium entry and cancer: updated views and insights. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2017; 46:395-413. [PMID: 28516266 DOI: 10.1007/s00249-017-1216-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 03/10/2017] [Accepted: 04/26/2017] [Indexed: 12/20/2022]
Abstract
Tight control of basal cytosolic Ca2+ concentration is essential for cell survival and to fine-tune Ca2+-dependent cell functions. A way to control this basal cytosolic Ca2+ concentration is to regulate membrane Ca2+ channels including store-operated Ca2+ channels and secondary messenger-operated channels linked to G-protein-coupled or tyrosine kinase receptor activation. Orai, with or without its reticular STIM partner and Transient Receptor Potential (TRP) proteins, were considered to be the main Ca2+ channels involved. It is well accepted that, in response to cell stimulation, opening of these Ca2+ channels contributes to Ca2+ entry and the transient increase in cytosolic Ca2+ concentration involved in intracellular signaling. However, in various experimental conditions, Ca2+ entry and/or Ca2+ currents can be recorded at rest, without application of any experimental stimulation. This led to the proposition that some plasma membrane Ca2+ channels are already open/activated in basal condition, contributing therefore to constitutive Ca2+ entry. This article focuses on direct and indirect observations supporting constitutive activity of channels belonging to the Orai and TRP families and on the mechanisms underlying their basal/constitutive activities.
Collapse
Affiliation(s)
- Olivier Mignen
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Bruno Constantin
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marie Potier-Cartereau
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aubin Penna
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Mathieu Gautier
- EA4667, Université de Picardie Jules Verne, 80039, Amiens, France
| | - Maxime Guéguinou
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Yves Renaudineau
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Kenji F Shoji
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Romain Félix
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Elsa Bayet
- STIM, ERL 7368 CNRS Université de Poitiers, Poitiers, France
- IRSET, Inserm U1085, University of Rennes 1, 36043, Rennes, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Paul Buscaglia
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Marjolaine Debant
- Inserm UMR 1078 IFR148 Université de Bretagne Occidentale, Brest, France
- EA 2216, Inserm ESPRI, ERI 29, Brest, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Aurélie Chantôme
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France
| | - Christophe Vandier
- Inserm/University of Tours U1069, Nutrition-Croissance et Cancer (N2C), 37032, Tours, France.
- Network "Ion Channels and Cancer-Canceropôle Grand Ouest", (IC-CGO), Grand Ouest, France.
| |
Collapse
|
32
|
Mitochondria mediate cell membrane repair and contribute to Duchenne muscular dystrophy. Cell Death Differ 2016; 24:330-342. [PMID: 27834955 PMCID: PMC5299714 DOI: 10.1038/cdd.2016.127] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/02/2016] [Accepted: 09/28/2016] [Indexed: 12/31/2022] Open
Abstract
Dystrophin deficiency is the genetic basis for Duchenne muscular dystrophy (DMD), but the cellular basis of progressive myofiber death in DMD is not fully understood. Using two dystrophin-deficient mdx mouse models, we find that the mitochondrial dysfunction is among the earliest cellular deficits of mdx muscles. Mitochondria in dystrophic myofibers also respond poorly to sarcolemmal injury. These mitochondrial deficits reduce the ability of dystrophic muscle cell membranes to repair and are associated with a compensatory increase in dysferlin-mediated membrane repair proteins. Dysferlin deficit in mdx mice further compromises myofiber cell membrane repair and enhances the muscle pathology at an asymptomatic age for dysferlin-deficient mice. Restoring partial dystrophin expression by exon skipping improves mitochondrial function and offers potential to improve myofiber repair. These findings identify that mitochondrial deficit in muscular dystrophy compromises the repair of injured myofibers and show that this repair mechanism is distinct from and complimentary to the dysferlin-mediated repair of injured myofibers.
Collapse
|
33
|
Iwata Y, Suzuki N, Ohtake H, Kamauchi S, Hashimoto N, Kiyono T, Wakabayashi S. Cancer cachexia causes skeletal muscle damage via transient receptor potential vanilloid 2-independent mechanisms, unlike muscular dystrophy. J Cachexia Sarcopenia Muscle 2016; 7:366-76. [PMID: 27239414 PMCID: PMC4864294 DOI: 10.1002/jcsm.12067] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 06/05/2015] [Accepted: 07/28/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Muscle wasting during cancer cachexia contributes to patient morbidity. Cachexia-induced muscle damage may be understood by comparing its symptoms with those of other skeletal muscle diseases, but currently available data are limited. METHODS We modelled cancer cachexia in mice bearing Lewis lung carcinoma/colon adenocarcinoma and compared the associated muscle damage with that in a murine muscular dystrophy model (mdx mice). We measured biochemical and immunochemical parameters: amounts/localization of cytoskeletal proteins and/or Ca(2+) signalling proteins related to muscle function and abnormality. We analysed intracellular Ca(2+) mobilization and compared results between the two models. Involvement of Ca(2+)-permeable channel transient receptor potential vanilloid 2 (TRPV2) was examined by inoculating Lewis lung carcinoma cells into transgenic mice expressing dominant-negative TRPV2. RESULTS Tumourigenesis caused loss of body and skeletal muscle weight and reduced muscle force and locomotor activity. Similar to mdx mice, cachexia muscles exhibited myolysis, reduced sarcolemmal sialic acid content, and enhanced lysosomal exocytosis and sarcolemmal localization of phosphorylated Ca(2+)/CaMKII. Abnormal autophagy and degradation of dystrophin also occurred. Unlike mdx muscles, cachexia muscles did not exhibit regeneration markers (centrally nucleated fibres), and levels of autophagic proteolytic pathway markers increased. While a slight accumulation of TRPV2 was observed in cachexia muscles, Ca(2+) influx via TRPV2 was not elevated in cachexia-associated myotubes, and the course of cachexia pathology was not ameliorated by dominant-negative inhibition of TRPV2. CONCLUSIONS Thus, cancer cachexia may induce muscle damage through TRPV2-independent mechanisms distinct from those in muscular dystrophy; this may help treat patients with tumour-induced muscle wasting.
Collapse
Affiliation(s)
- Yuko Iwata
- Department of Molecular Physiology National Cerebral and Cardiovascular Center Research Institute Suita Osaka 565-8565 Japan
| | | | - Hitomi Ohtake
- Department of Molecular Physiology National Cerebral and Cardiovascular Center Research Institute Suita Osaka 565-8565 Japan
| | - Shinya Kamauchi
- Department of Molecular Physiology National Cerebral and Cardiovascular Center Research Institute Suita Osaka 565-8565 Japan
| | - Naohiro Hashimoto
- Department of Regenerative Medicine, National Institute for Longevity Science National Center for Geriatrics and Gerontology Oobu Aichi 474-8522 Japan
| | - Tohru Kiyono
- Viology Division National Cancer Center Research Institute Chuo-ku Tokyo 104-0045 Japan
| | - Shigeo Wakabayashi
- Department of Molecular Physiology National Cerebral and Cardiovascular Center Research Institute Suita Osaka 565-8565 Japan
| |
Collapse
|
34
|
Stelter Z, Strakova J, Yellamilli A, Fischer K, Sharpe K, Townsend D. Hypoxia-induced cardiac injury in dystrophic mice. Am J Physiol Heart Circ Physiol 2016; 310:H938-48. [PMID: 26851247 DOI: 10.1152/ajpheart.00917.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/02/2016] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a disease of progressive destruction of striated muscle, resulting in muscle weakness with progressive respiratory and cardiac failure. Respiratory and cardiac disease are the leading causes of death in DMD patients. Previous studies have suggested an important link between cardiac dysfunction and hypoxia in the dystrophic heart; these studies aim to understand the mechanism underlying this connection. Here we demonstrate that anesthetized dystrophic mice display significant mortality following acute exposure to hypoxia. This increased mortality is associated with a significant metabolic acidosis, despite having significantly higher levels of arterial Po2 Chronic hypoxia does not result in mortality, but rather is characterized by marked cardiac fibrosis. Studies in isolated hearts reveal that the contractile function of dystrophic hearts is highly susceptible to short bouts of ischemia, but these hearts tolerate prolonged acidosis better than wild-type hearts, indicating an increased sensitivity of the dystrophic heart to hypoxia. Dystrophic hearts display decreased cardiac efficiency and oxygen extraction. Isolated dystrophic cardiomyocytes and hearts have normal levels of FCCP-induced oxygen consumption, and mitochondrial morphology and content are normal in the dystrophic heart. These studies demonstrate reductions in cardiac efficiency and oxygen extraction of the dystrophic heart. The underlying cause of this reduced oxygen extraction is not clear; however, the current studies suggest that large disruptions of mitochondrial respiratory function or coronary flow regulation are not responsible. This finding is significant, as hypoxia is a common and largely preventable component of DMD that may contribute to the progression of the cardiac disease in DMD patients.
Collapse
Affiliation(s)
- Zachary Stelter
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Jana Strakova
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Amritha Yellamilli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Kaleb Fischer
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Katharine Sharpe
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
35
|
Paran CW, Zou K, Ferrara PJ, Song H, Turk J, Funai K. Lipogenesis mitigates dysregulated sarcoplasmic reticulum calcium uptake in muscular dystrophy. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1530-8. [PMID: 26361872 DOI: 10.1016/j.bbalip.2015.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/27/2015] [Accepted: 09/06/2015] [Indexed: 01/07/2023]
Abstract
Muscular dystrophy is accompanied by a reduction in activity of sarco/endoplasmic reticulum Ca(2+)-ATPase (SERCA) that contributes to abnormal Ca(2+) homeostasis in sarco/endoplasmic reticulum (SR/ER). Recent findings suggest that skeletal muscle fatty acid synthase (FAS) modulates SERCA activity and muscle function via its effects on SR membrane phospholipids. In this study, we examined muscle's lipid metabolism in mdx mice, a mouse model for Duchenne muscular dystrophy (DMD). De novo lipogenesis was ~50% reduced in mdx muscles compared to wildtype (WT) muscles. Gene expressions of lipogenic and other ER lipid-modifying enzymes were found to be differentially expressed between wildtype (WT) and mdx muscles. A comprehensive examination of muscles' SR phospholipidome revealed elevated phosphatidylcholine (PC) and PC/phosphatidylethanolamine (PE) ratio in mdx compared to WT mice. Studies in primary myocytes suggested that defects in key lipogenic enzymes including FAS, stearoyl-CoA desaturase-1 (SCD1), and Lipin1 are likely contributing to reduced SERCA activity in mdx mice. Triple transgenic expression of FAS, SCD1, and Lipin1 (3TG) in mdx myocytes partly rescued SERCA activity, which coincided with an increase in SR PE that normalized PC/PE ratio. These findings implicate a defect in lipogenesis to be a contributing factor for SERCA dysfunction in muscular dystrophy. Restoration of muscle's lipogenic pathway appears to mitigate SERCA function through its effects on SR membrane composition.
Collapse
Affiliation(s)
- Christopher W Paran
- Department of Kinesiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; Department of Physiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA
| | - Kai Zou
- Department of Kinesiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA
| | - Patrick J Ferrara
- Department of Kinesiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA
| | - Haowei Song
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | - John Turk
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | - Katsuhiko Funai
- Department of Kinesiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; Department of Physiology, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, 115 Heart Drive, Greenville, North Carolina 27834, USA.
| |
Collapse
|
36
|
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
|
37
|
Guiraud S, Squire SE, Edwards B, Chen H, Burns DT, Shah N, Babbs A, Davies SG, Wynne GM, Russell AJ, Elsey D, Wilson FX, Tinsley JM, Davies KE. Second-generation compound for the modulation of utrophin in the therapy of DMD. Hum Mol Genet 2015; 24:4212-24. [PMID: 25935002 PMCID: PMC4492389 DOI: 10.1093/hmg/ddv154] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/27/2015] [Indexed: 01/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. There is currently no cure for DMD although various promising approaches are progressing through human clinical trials. By pharmacologically modulating the expression of the dystrophin-related protein utrophin, we have previously demonstrated in dystrophin-deficient mdx studies, daily SMT C1100 treatment significantly reduced muscle degeneration leading to improved muscle function. This manuscript describes the significant disease modifying benefits associated with daily dosing of SMT022357, a second-generation compound in this drug series with improved physicochemical properties and a more robust metabolism profile. These studies in the mdx mouse demonstrate that oral administration of SMT022357 leads to increased utrophin expression in skeletal, respiratory and cardiac muscles. Significantly, utrophin expression is localized along the length of the muscle fibre, not just at the synapse, and is fibre-type independent, suggesting that drug treatment is modulating utrophin transcription in extra-synaptic myonuclei. This results in improved sarcolemmal stability and prevents dystrophic pathology through a significant reduction of regeneration, necrosis and fibrosis. All these improvements combine to protect the mdx muscle from contraction induced damage and enhance physiological function. This detailed evaluation of the SMT C1100 drug series strongly endorses the therapeutic potential of utrophin modulation as a disease modifying therapeutic strategy for all DMD patients irrespective of their dystrophin mutation.
Collapse
Affiliation(s)
- Simon Guiraud
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK,
| | - Sarah E Squire
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Benjamin Edwards
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Huijia Chen
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - David T Burns
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Nandini Shah
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Arran Babbs
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Stephen G Davies
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Graham M Wynne
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Angela J Russell
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3PT, UK and
| | - David Elsey
- Summit Therapeutics plc, 85b Park Drive, Milton Park, Abingdon, Oxfordshire OX14 4RY, UK
| | - Francis X Wilson
- Summit Therapeutics plc, 85b Park Drive, Milton Park, Abingdon, Oxfordshire OX14 4RY, UK
| | - Jon M Tinsley
- Summit Therapeutics plc, 85b Park Drive, Milton Park, Abingdon, Oxfordshire OX14 4RY, UK
| | - Kay E Davies
- Medical Research Council Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK,
| |
Collapse
|
38
|
Holland A, Dowling P, Meleady P, Henry M, Zweyer M, Mundegar RR, Swandulla D, Ohlendieck K. Label-free mass spectrometric analysis of the mdx-4cv diaphragm identifies the matricellular protein periostin as a potential factor involved in dystrophinopathy-related fibrosis. Proteomics 2015; 15:2318-31. [PMID: 25737063 DOI: 10.1002/pmic.201400471] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/19/2014] [Accepted: 02/26/2015] [Indexed: 01/17/2023]
Abstract
Proteomic profiling plays a decisive role in the identification of novel biomarkers of muscular dystrophy and the elucidation of new pathobiochemical mechanisms that underlie progressive muscle wasting. Building on the findings of recent comparative analyses of tissue samples and body fluids from dystrophic animals and patients afflicted with Duchenne muscular dystrophy, we have used here label-free MS to study the severely dystrophic diaphragm from the not extensively characterized mdx-4cv mouse. This animal model of progressive muscle wasting exhibits less dystrophin-positive revertant fibers than the conventional mdx mouse, making it ideal for the future monitoring of experimental therapies. The pathoproteomic signature of the mdx-4cv diaphragm included a significant increase in the fibrosis marker collagen and related extracellular matrix proteins (asporin, decorin, dermatopontin, prolargin) and cytoskeletal proteins (desmin, filamin, obscurin, plectin, spectrin, tubulin, vimentin, vinculin), as well as decreases in proteins of ion homeostasis (parvalbumin) and the contractile apparatus (myosin-binding protein). Importantly, one of the most substantially increased proteins was identified as periostin, a matricellular component and apparent marker of fibrosis and tissue damage. Immunoblotting confirmed a considerable increase of periostin in the dystrophin-deficient diaphragm from both mdx and mdx-4cv mice, suggesting an involvement of this matricellular protein in dystrophinopathy-related fibrosis.
Collapse
Affiliation(s)
- Ashling Holland
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, County Kildare, Ireland
| | - Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, County Kildare, Ireland
| | - Paula Meleady
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Michael Henry
- National Institute for Cellular Biotechnology, Dublin City University, Dublin, Ireland
| | - Margit Zweyer
- Department of Physiology II, University of Bonn, Bonn, Germany
| | | | | | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, County Kildare, Ireland
| |
Collapse
|
39
|
Manning J, O'Malley D. What has the mdx mouse model of Duchenne muscular dystrophy contributed to our understanding of this disease? J Muscle Res Cell Motil 2015; 36:155-67. [PMID: 25669899 DOI: 10.1007/s10974-015-9406-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/28/2015] [Indexed: 12/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-chromosome linked recessive disorder caused by the truncation or deletion of the dystrophin gene. The most widely used animal model of this disease is the dystrophin-deficient mdx mouse which was first discovered 30 years ago. Despite its extensive use in DMD research, no effective treatment has yet been developed for this devastating disease. This review explores what we have learned from this mouse model regarding the pathophysiology of DMD and asks if it has a future in providing a better more thorough understanding of this disease or if it will bring us any closer to improving the outlook for DMD patients.
Collapse
Affiliation(s)
- Jennifer Manning
- Department of Physiology, University College Cork, 4.23 Western Gateway Building, Cork, Ireland
| | | |
Collapse
|
40
|
Regional circumferential strain is a biomarker for disease severity in duchenne muscular dystrophy heart disease: a cross-sectional study. Pediatr Cardiol 2015; 36:111-9. [PMID: 25085262 DOI: 10.1007/s00246-014-0972-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/16/2014] [Indexed: 10/25/2022]
Abstract
The aim of this study is to determine the contribution of strain ε cc in mid left ventricular (LV) segments to the reduction of composite LV circumferential ε cc in assess severity of duchenne muscular dystrophy (DMD) heart disease as assessed by cardiac magnetic resonance imaging (CMR). DMD patients and control subjects were stratified by age, LV ejection fraction, and late gadolinium enhancement (LGE) status. Tagged CMR images were analyzed for global ventricular function, LGE imaging, and composite and segmental ε cc. The relationship between changes in segmental ε cc changes and LGE across patient groups was assessed by a statistical step-down model. LV ε cc exhibited segmental heterogeneity; in control subjects and young DMD patients, ε cc was greatest in LV lateral free wall segments. However, with increasing age and cardiac disease severity as demonstrated by decreased EF and development of myocardial strain the segmental differences diminished. In subjects with advanced heart disease as evidenced by reduced LV ejection fraction and presence of LGE, very little segmental heterogeneity was present. In control subjects and young DMD patients, ε cc was greatest in LV lateral free wall segments. Increased DMD heart disease severity was associated with reduced composite; ε cc diminished regional ε cc heterogeneity and positive LGE imaging. Taken together, these findings suggest that perturbation of segmental, heterogeneous ε cc is an early biomarker of disease severity in this cross-section of DMD patients.
Collapse
|
41
|
Rybalka E, Timpani CA, Cooke MB, Williams AD, Hayes A. Defects in mitochondrial ATP synthesis in dystrophin-deficient mdx skeletal muscles may be caused by complex I insufficiency. PLoS One 2014; 9:e115763. [PMID: 25541951 PMCID: PMC4277356 DOI: 10.1371/journal.pone.0115763] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/01/2014] [Indexed: 12/21/2022] Open
Abstract
Duchenne Muscular Dystrophy is a chronic, progressive and ultimately fatal skeletal muscle wasting disease characterised by sarcolemmal fragility and intracellular Ca2+ dysregulation secondary to the absence of dystrophin. Mounting literature also suggests that the dysfunction of key energy systems within the muscle may contribute to pathological muscle wasting by reducing ATP availability to Ca2+ regulation and fibre regeneration. No study to date has biochemically quantified and contrasted mitochondrial ATP production capacity by dystrophic mitochondria isolated from their pathophysiological environment such to determine whether mitochondria are indeed capable of meeting this heightened cellular ATP demand, or examined the effects of an increasing extramitochondrial Ca2+ environment. Using isolated mitochondria from the diaphragm and tibialis anterior of 12 week-old dystrophin-deficient mdx and healthy control mice (C57BL10/ScSn) we have demonstrated severely depressed Complex I-mediated mitochondrial ATP production rate in mdx mitochondria that occurs irrespective of the macronutrient-derivative substrate combination fed into the Kreb's cycle, and, which is partially, but significantly, ameliorated by inhibition of Complex I with rotenone and stimulation of Complex II-mediated ATP-production with succinate. There was no difference in the MAPR response of mdx mitochondria to increasing extramitochondrial Ca2+ load in comparison to controls, and 400 nM extramitochondrial Ca2+ was generally shown to be inhibitory to MAPR in both groups. Our data suggests that DMD pathology is exacerbated by a Complex I deficiency, which may contribute in part to the severe reductions in ATP production previously observed in dystrophic skeletal muscle.
Collapse
Affiliation(s)
- Emma Rybalka
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Western Health, Victoria, Australia
| | - Cara A. Timpani
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Matthew B. Cooke
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Western Health, Victoria, Australia
| | - Andrew D. Williams
- School of Human Life Sciences, University of Tasmania, Launceston, Australia
| | - Alan Hayes
- Centre for Chronic Disease Prevention and Management, College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
- Institute of Sport, Exercise & Active Living (ISEAL), Victoria University, Melbourne, Victoria, Australia
- Australian Institute of Musculoskeletal Science, Western Health, Victoria, Australia
| |
Collapse
|
42
|
Abstract
Muscular dystrophies are a group of diseases characterised by the primary wasting of skeletal muscle, which compromises patient mobility and in the most severe cases originate a complete paralysis and premature death. Existing evidence implicates calcium dysregulation as an underlying crucial event in the pathophysiology of several muscular dystrophies, such as dystrophinopathies, calpainopathies or myotonic dystrophy among others. Duchenne muscular dystrophy is the most frequent myopathy in childhood, and calpainopathy or LGMD2A is the most common form of limb-girdle muscular dystrophy, whereas myotonic dystrophy is the most frequent inherited muscle disease worldwide. In this review, we summarise recent advances in our understanding of calcium ion cycling through the sarcolemma, the sarcoplasmic reticulum and mitochondria, and its involvement in the pathogenesis of these dystrophies. We also discuss some of the clinical implications of recent findings regarding Ca2+ handling as well as novel approaches to treat muscular dystrophies targeting Ca2+ regulatory proteins.
Collapse
|
43
|
|
44
|
Uaesoontrachoon K, Quinn JL, Tatem KS, Van Der Meulen JH, Yu Q, Phadke A, Miller BK, Gordish-Dressman H, Ongini E, Miglietta D, Nagaraju K. Long-term treatment with naproxcinod significantly improves skeletal and cardiac disease phenotype in the mdx mouse model of dystrophy. Hum Mol Genet 2014; 23:3239-49. [PMID: 24463621 DOI: 10.1093/hmg/ddu033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In Duchenne muscular dystrophy (DMD) patients and the mouse model of DMD, mdx, dystrophin deficiency causes a decrease and mislocalization of muscle-specific neuronal nitric oxide synthase (nNOSμ), leading to functional impairments. Previous studies have shown that nitric oxide (NO) donation associated with anti-inflammatory action has beneficial effects in dystrophic mouse models. In this study, we have systematically investigated the effects of naproxcinod, an NO-donating naproxen derivative, on the skeletal and cardiac disease phenotype in mdx mice. Four-week-old mdx and C57BL/10 mice were treated with four different concentrations (0, 10, 21 and 41 mg/kg) of naproxcinod and 0.9 mg/kg of prednisolone in their food for 9 months. All mice were subjected to twice-weekly treadmill sessions, and functional and behavioral parameters were measured at 3, 6 and 9 months of treatment. In addition, we evaluated in vitro force contraction, optical imaging of inflammation, echocardiography and blood pressure (BP) at the 9-month endpoint prior to sacrifice. We found that naproxcinod treatment at 21 mg/kg resulted in significant improvement in hindlimb grip strength and a 30% decrease in inflammation in the fore- and hindlimbs of mdx mice. Furthermore, we found significant improvement in heart function, as evidenced by improved fraction shortening, ejection fraction and systolic BP. In addition, the long-term detrimental effects of prednisolone typically seen in mdx skeletal and heart function were not observed at the effective dose of naproxcinod. In conclusion, our results indicate that naproxcinod has significant potential as a safe therapeutic option for the treatment of muscular dystrophies.
Collapse
Affiliation(s)
| | - James L Quinn
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Kathleen S Tatem
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Jack H Van Der Meulen
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Qing Yu
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Aditi Phadke
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Brittany K Miller
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | - Heather Gordish-Dressman
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Ennio Ongini
- Nicox Research Institute, Via Ariosto 21-20091, Bresso Milano, Italy
| | - Daniela Miglietta
- Nicox Research Institute, Via Ariosto 21-20091, Bresso Milano, Italy
| | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
45
|
Hor KN, Taylor MD, Al-Khalidi HR, Cripe LH, Raman SV, Jefferies JL, O’Donnell R, Benson DW, Mazur W. Prevalence and distribution of late gadolinium enhancement in a large population of patients with Duchenne muscular dystrophy: effect of age and left ventricular systolic function. J Cardiovasc Magn Reson 2013; 15:107. [PMID: 24359596 PMCID: PMC3896985 DOI: 10.1186/1532-429x-15-107] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/13/2013] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD), an X-linked disorder affects approximately 1 in 5000 males, is universally associated with heart disease. We previously identified myocardial disease by late gadolinium enhancement (LGE) in DMD subjects at various stages of disease, but the true prevalence is unclear. Cardiovascular magnetic resonance (CMR) is well established for both assessment of ventricular function and myocardial fibrosis by LGE. We sought to establish i) prevalence and distribution of LGE in a large DMD population and ii) relationship among LGE, age, LVEF by CMR and current living status. METHODS Current living status, demographic and CMR data including ventricular volumes, LVEF and LGE from 314 DMD patients undergoing evaluation at a single large tertiary referral center were analyzed. RESULTS 113 of 314 (36%) of DMD subjects showed LGE positivity with prevalence increasing from 17% of patients <10 years to 34% of those aged 10-15 years and 59% of those >15 years-old. Patients with LVEF ≥55% were LGE positive in 30% of cases; this increased to 84% for LVEF <55%. LGE was more prevalent in the free wall (531/1243, 42.7%) vs. septal segments (30/565, 5.3%). Patients with septal involvement were significantly older and had lower LVEF than those with isolated free wall LGE. Ten percent (11/113) patients who had LGE died 10.8 months after CMR. Only one patient from the LGE negative group died. Patients who died had higher heart rate, larger left ventricular volume and mass, greater number of positive LGE segment and increase incident of septal LGE compared to those who remained alive. CONCLUSION In DMD patients, LGE occurs early, is progressive and increases with both age and decreasing LVEF. Segmentally, the incidence of the number of positive LGE segments increase with age and lower LVEF. Older patients and those who died during the study period had more septal LGE involvement. The current studies suggest that the time course and distribution of LGE-positivity may be an important clinical biomarker to aid in the management of DMD-associated cardiac disease.
Collapse
Affiliation(s)
- Kan N Hor
- Nationwide Children’s Hospital, Columbus, OH, USA
| | | | | | | | | | | | | | | | - Wojciech Mazur
- The Heart and Vascular Center at the Christ Hospital, Cincinnati, OH, USA
| |
Collapse
|
46
|
Moloughney JG, Weisleder N. Poloxamer 188 (p188) as a membrane resealing reagent in biomedical applications. Recent Pat Biotechnol 2013; 6:200-11. [PMID: 23092436 DOI: 10.2174/1872208311206030200] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/30/2012] [Accepted: 08/04/2012] [Indexed: 11/22/2022]
Abstract
Maintenance of the integrity of the plasma membrane is essential for maintenance of cellular function and prevention of cell death. Since the plasma membrane is frequently exposed to a variety of mechanical and chemical insults the cell has evolved active processes to defend against these injuries by resealing disruptions in the plasma membrane. Cell membrane repair is a conserved process observed in nearly every cell type where intracellular vesicles are recruited to sites of membrane disruption where they can fuse with themselves or the plasma membrane to create a repair patch. When disruptions are extensive or there is an underlying pathology that reduces the membrane repair capacity of a cell this defense mechanism may prove insufficient and the cell could die due to breakdown of the plasma membrane. Extensive loss of cells can compromise the integrity and function of tissues and leading to disease. Thus, methods to increase membrane resealing capacity could have broad utility in a number of disease states. Efforts to find reagents that can modulate plasma membrane reseal found that specific tri-block copolymers, such as poloxamer 188 (P188, or Pluronic F68), can increase the structural stability and resealing of the plasma membrane. Here we review several current patents and patent applications that present inventions making use of P188 and other copolymers to treat specific disease states such as muscular dystrophy, heart failure, neurodegenerative disorders and electrical injuries, or to facilitate biomedical applications such as transplantation. There appears to be promise for the application of poloxamers in the treatment of various diseases, however there are potential concerns with toxicity with long term application and bioavailability in some cases.
Collapse
Affiliation(s)
- Joseph G Moloughney
- Department of Neuroscience and Cell Biology, UMDNJ- Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | |
Collapse
|
47
|
Giliberto F, Ferreiro V, Dalamon V, Szijan I. Dystrophin deletions and cognitive impairment in Duchenne/Becker muscular dystrophy. Neurol Res 2013; 26:83-7. [PMID: 14977063 DOI: 10.1179/016164104773026589] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Analyses of deletions in the dystrophin gene and of cognitive status were performed on patients with Duchenne (DMD) or Becker (BMD) muscular dystrophy in order to find a correlation between both features. Molecular study by multiplex and simplex PCR of dystrophin exons led to the identification of 51 deletions in 126 unrelated patients. Most of them were frameshift, in full agreement with severe clinical symptoms, three patients with a BMD-like phenotype had in-frame mutations. Deletions were localized with reference to the different dystrophin isoform sequences and were clustered in two main areas, 5' and central+ 3' end of the gene. Cognitive abilities were tested in 47 out of 51 patients with identified mutations, 23 of them being mentally impaired. Comparison of molecular and neuropsychological features showed that deletions localized in central and 3' parts of the gene (18 out of 23) are preferentially associated with mental impairment. Fourteen of them were found in the regulatory and coding sequences for the three CNS specific carboxy terminal isoforms. Therefore, though mutations with variable locations may lead to cognitive impairment, our results show that deletions in the distal portion of the gene are basically related to mental retardation.
Collapse
Affiliation(s)
- Florencia Giliberto
- Genetica y Biologia Molecular, Facultad de Farmacia y Bioquímica, Instituto de Neurociencias Aplicadas del Hospital de Clinicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | | | | |
Collapse
|
48
|
Shin J, Tajrishi MM, Ogura Y, Kumar A. Wasting mechanisms in muscular dystrophy. Int J Biochem Cell Biol 2013; 45:2266-79. [PMID: 23669245 DOI: 10.1016/j.biocel.2013.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022]
Abstract
Muscular dystrophy is a group of more than 30 different clinical genetic disorders that are characterized by progressive skeletal muscle wasting and degeneration. Primary deficiency of specific extracellular matrix, sarcoplasmic, cytoskeletal, or nuclear membrane protein results in several secondary changes such as sarcolemmal instability, calcium influx, fiber necrosis, oxidative stress, inflammatory response, breakdown of extracellular matrix, and eventually fibrosis which leads to loss of ambulance and cardiac and respiratory failure. A number of molecular processes have now been identified which hasten disease progression in human patients and animal models of muscular dystrophy. Accumulating evidence further suggests that aberrant activation of several signaling pathways aggravate pathological cascades in dystrophic muscle. Although replacement of defective gene with wild-type is paramount to cure, management of secondary pathological changes has enormous potential to improving the quality of life and extending lifespan of muscular dystrophy patients. In this article, we have reviewed major cellular and molecular mechanisms leading to muscle wasting in muscular dystrophy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Jonghyun Shin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
49
|
Harisseh R, Chatelier A, Magaud C, Déliot N, Constantin B. Involvement of TRPV2 and SOCE in calcium influx disorder in DMD primary human myotubes with a specific contribution of α1-syntrophin and PLC/PKC in SOCE regulation. Am J Physiol Cell Physiol 2013; 304:C881-94. [DOI: 10.1152/ajpcell.00182.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Calcium homeostasis is critical for several vital functions in excitable and nonexcitable cells and has been shown to be impaired in many pathologies including Duchenne muscular dystrophy (DMD). Various studies using murine models showed the implication of calcium entry in the dystrophic phenotype. However, alteration of store-operated calcium entry (SOCE) and transient receptor potential vanilloid 2 (TRPV2)-dependant cation entry has not been investigated yet in human skeletal muscle cells. We pharmacologically characterized basal and store-operated cation entries in primary cultures of myotubes prepared from muscle of normal and DMD patients and found, for the first time, an increased SOCE in DMD myotubes. Moreover, this increase cannot be explained by an over expression of the well-known SOCE actors: TRPC1/4, Orai1, and stromal interaction molecule 1 (STIM1) mRNA and proteins. Thus we investigated the modes of regulation of this cation entry. We firstly demonstrated the important role of the scaffolding protein α1-syntrophin, which regulates SOCE in primary human myotubes through its PDZ domain. We also studied the implication of phospholipase C (PLC) and protein kinase C (PKC) in SOCE and showed that their inhibition restores normal levels of SOCE in DMD human myotubes. In addition, the involvement of TRPV2 in calcium deregulation in DMD human myotubes was explored. We showed an abnormal elevation of TRPV2-dependant cation entry in dystrophic primary human myotubes compared with normal ones. These findings show that calcium homeostasis mishandling in DMD myotubes depends on SOCE under the influence of Ca2+/PLC/PKC pathway and α1-syntrophin regulation as well as on TRPV2-dependant cation influx.
Collapse
Affiliation(s)
- Rania Harisseh
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Aurélien Chatelier
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Christophe Magaud
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Nadine Déliot
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| | - Bruno Constantin
- Institut de Physiologie et Biologie Cellulaires, Université de Poitiers/Centre National de la Recherche Scientifique FRE-3511 Poitiers, France
| |
Collapse
|
50
|
Mosqueira M, Zeiger U, Förderer M, Brinkmeier H, Fink RHA. Cardiac and respiratory dysfunction in Duchenne muscular dystrophy and the role of second messengers. Med Res Rev 2013; 33:1174-213. [PMID: 23633235 DOI: 10.1002/med.21279] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Duchenne muscular dystrophy (DMD) affects young boys and is characterized by the absence of dystrophin, a large cytoskeletal protein present in skeletal and cardiac muscle cells and neurons. The heart and diaphragm become necrotic in DMD patients and animal models of DMD, resulting in cardiorespiratory failure as the leading cause of death. The major consequences of the absence of dystrophin are high levels of intracellular Ca(2+) and the unbalanced production of NO that can finally trigger protein degradation and cell death. Cytoplasmic increase in Ca(2+) concentration directly and indirectly triggers different processes such as necrosis, fibrosis, and activation of macrophages. The absence of the neuronal isoform of nitric oxide synthase (nNOS) and the overproduction of NO by the inducible isoform (iNOS) further increase the intracellular Ca(2+) via a hypernitrosylation of the ryanodine receptor. NO overproduction, which further induces the expression of iNOS but decreases the expression of the endothelial isoform (eNOS), deregulates the muscle tissue blood flow creating an ischemic situation. The high levels of Ca(2+) in dystrophic muscles and the ischemic state of the muscle tissue would culminate in a positive feedback loop. While efforts continue toward optimizing cardiac and respiratory care of DMD patients, both Ca(2+) and NO in cardiac and respiratory muscle pathways have been shown to be important to the etiology of the disease. Understanding the mechanisms behind the fine regulation of Ca(2+) -NO may be important for a noninterventional and noninvasive supportive approach to treat DMD patients, improving the quality of life and natural history of DMD patients.
Collapse
Affiliation(s)
- Matias Mosqueira
- Medical Biophysics Unit, Institute of Physiology and Pathophysiology, INF326, Heidelberg University, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|