1
|
Hu X, McCrady AN, Bukovec KE, Yuan C, Miller EY, Bour RK, Bruce AC, Crump KB, Peirce SM, Grange RW, Blemker SS. A novel ex vivo protocol that mimics length and excitation changes of human muscles during walking induces force losses in EDL but not in soleus of mdx mice. PLoS One 2025; 20:e0320901. [PMID: 40193354 PMCID: PMC11975108 DOI: 10.1371/journal.pone.0320901] [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: 09/17/2024] [Accepted: 02/26/2025] [Indexed: 04/09/2025] Open
Abstract
Although eccentric contraction protocols are widely used to study the pathophysiology and potential treatments for Duchenne muscular dystrophy (DMD), they do not reflect the stresses, strains, strain rates, and excitation profiles that DMD muscles experience during human daily functional tasks, like walking. This limitation of eccentric contractions may impede our understanding of disease progression in DMD and proper assessment of treatment efficacy. The goals of this study were to examine the extent of force loss induced by a gait cycling protocol we developed, and compare to that from a typical eccentric contraction protocol in soleus and extensor digitorum longus (EDL) muscles of mdx mice. To achieve this goal, mdx soleus and EDL muscles were subjected to eccentric contractions at three levels of strain (10%, 20% and 30% optimal length Lo) and up to 200 cycles of our gait cycling protocol that mimicked the length changes and excitation patterns of the corresponding muscles during human walking gait. Our results showed that EDL but not soleus muscles had significant losses in isometric tetanic forces after the cycling protocols. Compared to the eccentric contraction protocol, the decrements in contractile performance from the cycling protocol were similar to those from the eccentric contractions at 10% in soleus and 20% Lo in EDL. Together, these results indicated the gait cycling protocol is a valuable experimental approach to better understand disease progression and to screen and evaluate efficacy of novel therapeutics for DMD.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Allison N. McCrady
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Katherine E. Bukovec
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Claire Yuan
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Emily Y. Miller
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Rachel K. Bour
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Anthony C. Bruce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Katherine B. Crump
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Shayn M. Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| | - Robert W. Grange
- Department of Human Nutrition, Foods, and Exercise and Metabolism Core, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Silvia S. Blemker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Orthopedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
2
|
Tiper Y, Xie Z, Hofemeier A, Lad H, Luber M, Krawetz R, Betz T, Zimmermann WH, Morton AB, Segal SS, Gilbert PM. Optimizing electrical field stimulation parameters reveals the maximum contractile function of human skeletal muscle microtissues. Am J Physiol Cell Physiol 2025; 328:C1160-C1176. [PMID: 40019026 DOI: 10.1152/ajpcell.00308.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/07/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Skeletal muscle microtissues are engineered to develop therapies for restoring muscle function in patients. However, optimal electrical field stimulation (EFS) parameters to evaluate the function of muscle microtissues remain unestablished. This study reports a protocol to optimize EFS parameters for eliciting contractile force of muscle microtissues cultured in micropost platforms. Muscle microtissues were produced across an opposing pair of microposts in polydimethylsiloxane and polymethyl methacrylate culture platforms using primary, immortalized, and induced pluripotent stem cell-derived myoblasts. In response to EFS between needle electrodes, contraction deflects microposts proportional to developed force. At 5 V, pulse durations used for native muscle (0.1-1 ms) failed to elicit contraction of microtissues; durations reported for engineered muscle (5-10 ms) failed to elicit peak force. Instead, pulse durations of 20-80 ms were required to elicit peak twitch force across microtissues derived from five myoblast lines. Similarly, although peak tetanic force occurs at 20-50 Hz for native human muscles, it varied across microtissues depending on the cell line type, ranging from 7 to 60 Hz. A new parameter, the dynamic oscillation of force, captured trends during rhythmic contractions, whereas quantifying the duration-at-peak force provides an extended kinetics parameter. Our findings indicate that muscle microtissues have cell line type-specific contractile properties, yet all contract and relax more slowly than native muscle, implicating underdeveloped excitation-contraction coupling. Failure to optimize EFS parameters can mask the functional potential of muscle microtissues by underestimating force production. Optimizing and reporting EFS parameters and metrics is necessary to leverage muscle microtissues for advancing skeletal muscle therapies.NEW & NOTEWORTHY Electrical field stimulation (EFS) parameters remain to be standardized for engineered skeletal muscle. Herein, we report a protocol for defining EFS parameters that elicit the maximal contractile force of muscle microtissues cultivated in micropost devices and highlight the value of developing appropriate metrics. The dynamic oscillation of force and duration-at-peak force are introduced as novel metrics of contraction kinetics.
Collapse
Affiliation(s)
- Yekaterina Tiper
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Zhuoye Xie
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Arne Hofemeier
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Lower Saxony, Göttingen, Germany
| | - Heta Lad
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Mattias Luber
- Third Institute of Physics, University of Göttingen, Göttingen, Germany
| | - Roman Krawetz
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Alberta, Canada
- Cummings School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Timo Betz
- Third Institute of Physics, University of Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner site Lower Saxony, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- German Center for Child and Adolescent Health (DZKJ), Göttingen, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, Göttingen, Germany
| | - Aaron B Morton
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, Texas, United States
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, Missouri, United States
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, United States
| | - Penney M Gilbert
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Hernández Rodríguez MY, Biswas DD, Slyne AD, Lee J, Scarrow E, Abdelbarr SM, Daniels H, O’Halloran KD, Ferreira LF, Gersbach CA, ElMallah MK. Respiratory pathology in the mdx/utrn -/- mouse: A murine model for Duchenne Muscular Dystrophy (DMD). PLoS One 2025; 20:e0316295. [PMID: 39919154 PMCID: PMC11805407 DOI: 10.1371/journal.pone.0316295] [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: 08/26/2024] [Accepted: 12/09/2024] [Indexed: 02/09/2025] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked devastating disease caused by a lack of dystrophin which results in progressive muscle weakness. As muscle weakness progresses, respiratory insufficiency and hypoventilation result in significant morbidity and mortality. The most studied DMD mouse model- the mdx mouse- has a milder respiratory phenotype compared to humans, likely due to compensatory overexpression of utrophin. mdx/utrn-/- mice lack both dystrophin and utrophin proteins. These mice have an early onset of muscular dystrophy, severe muscle weakness, and premature death, but the respiratory pathophysiology is unclear. The objective of this study is to characterize the respiratory pathophysiology and histopathology using whole body plethysmography to measure breathing and metabolism, diaphragm muscle functional analysis, histology, and immunohistochemistry. The mdx/utrn-/- mice have significant respiratory and metabolic deficits with respiratory insufficiency and hypoventilation when exposed to hypoxia and hypercarbia as early as 6 weeks of age. They also have significant diaphragmatic weakness and disrupted diaphragmatic structural pathology. The mdx/utrn-/- mice display respiratory dysfunction that mimics the DMD phenotype and therefore can provide a useful model to study the impact of novel therapies on respiratory function for DMD.
Collapse
Affiliation(s)
- Marán Y. Hernández Rodríguez
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Debolina D. Biswas
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Aoife D. Slyne
- Department of Physiology, University College Cork, Cork, Ireland
| | - Jane Lee
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Evelyn Scarrow
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Sarra M. Abdelbarr
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
| | - Heather Daniels
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
| | | | - Leonardo F. Ferreira
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina, United States of America
| | - Charles A. Gersbach
- Center for Advanced Genomic Technologies, Duke University, Durham, North Carolina, United States of America
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
- Department of Surgery, Duke University Hospital, Durham, North Carolina, United States of America
| | - Mai K. ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University, Durham, North Carolina, United States of America
- Department of Neurobiology, Duke University, Durham, North Carolina, United States of America
- Department of Cell Biology, Duke University, Durham, North Carolina, United States of America
| |
Collapse
|
4
|
Li Y, Li C, Sun Q, Liu X, Chen F, Cheung Y, Zhao Y, Xie T, Chazaud B, Sun H, Wang H. Skeletal muscle stem cells modulate niche function in Duchenne muscular dystrophy mouse through YY1-CCL5 axis. Nat Commun 2025; 16:1324. [PMID: 39900599 PMCID: PMC11790879 DOI: 10.1038/s41467-025-56474-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
Adult skeletal muscle stem cells (MuSCs) are indispensable for muscle regeneration and tightly regulated by macrophages (MPs) and fibro-adipogenic progenitors (FAPs) in their niche. Deregulated MuSC/MP/FAP interactions and the ensuing inflammation and fibrosis are hallmarks of dystrophic muscle. Here we demonstrate intrinsic deletion of transcription factor Yin Yang 1 (YY1) in MuSCs exacerbates dystrophic pathologies by altering composition and heterogeneity of MPs and FAPs. Further analysis reveals YY1 loss induces expression of immune genes in MuSCs, including C-C motif chemokine ligand 5 (Ccl5). Augmented CCL5 secretion promotes MP recruitment via CCL5/C-C chemokine receptor 5 (CCR5) crosstalk, which subsequently hinders FAP clearance through elevated Transforming growth factor-β1 (TGFβ1). Maraviroc-mediated pharmacological blockade of the CCL5/CCR5 axis effectively mitigates muscle dystrophy and improves muscle performance. Lastly, we demonstrate YY1 represses Ccl5 transcription by binding to its enhancer thus facilitating promoter-enhancer looping. Altogether, our study demonstrates the critical role of MuSCs in actively shaping their niche and provides novel insight into the therapeutic intervention of muscle dystrophy.
Collapse
MESH Headings
- Animals
- YY1 Transcription Factor/metabolism
- YY1 Transcription Factor/genetics
- Chemokine CCL5/metabolism
- Chemokine CCL5/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Mice
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Macrophages/metabolism
- Receptors, CCR5/metabolism
- Receptors, CCR5/genetics
- Mice, Inbred mdx
- Stem Cell Niche
- Mice, Knockout
- Maraviroc/pharmacology
- Mice, Inbred C57BL
- Male
- Transforming Growth Factor beta1/metabolism
- Stem Cells/metabolism
- Disease Models, Animal
Collapse
Grants
- 82172436 National Natural Science Foundation of China (National Science Foundation of China)
- 14115319, 14100620, 14106521, 14105823, 14120420, 14103522, 14105123 Research Grants Council, University Grants Committee (RGC, UGC)
- T13-602/21-N Research Grants Council, University Grants Committee (RGC, UGC)
- C6018-19GF Research Grants Council, University Grants Committee (RGC, UGC)
- 10210906, 08190626 Research Grants Council, University Grants Committee (RGC, UGC)
- AoE/M-402/20 Research Grants Council, University Grants Committee (RGC, UGC)
- STG1/E-403/24-N Research Grants Council, University Grants Committee (RGC, UGC)
- National Key R&D Program of China to H.W. (2022YFA0806003) Health and Medical Research Fund (HMRF) from Health Bureau of the Hong Kong Special Administrative Region, China to H.W. (10210906 and 08190626)
Collapse
Affiliation(s)
- Yang Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Chuhan Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qiang Sun
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China
| | - Xingyuan Liu
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fengyuan Chen
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yeelo Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Zhao
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ting Xie
- Center for Tissue Regeneration and Engineering, Division of Life Science, Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Bénédicte Chazaud
- Unité Physiopathologie et Génétique du Neurone et du Muscle, UMR CNRS 5261, Inserm U1315, Université Claude Bernard Lyon 1, Lyon, France
| | - Hao Sun
- Warshel Institute for Computational Biology, Faculty of Medicine, Chinese University of Hong Kong (Shenzhen), Guangdong, China.
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong SAR, China.
- Center for Neuromusculoskeletal Restorative Medicine Limited, Hong Kong Science Park, Hong Kong SAR, China.
| |
Collapse
|
5
|
Moretti A, Kupatt C, Wolf E. Cellular pathophysiology of Duchenne muscular dystrophy: insights from a novel rhesus macaque model. Signal Transduct Target Ther 2024; 9:357. [PMID: 39643645 PMCID: PMC11624258 DOI: 10.1038/s41392-024-02061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/09/2024] Open
Affiliation(s)
- Alessandra Moretti
- Medical Department I, Cardiology, Angiology, Pneumology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, 81675, Munich, Germany
- DZHK (German Center of Cardiovascular Research), Munich Heart Alliance, 80336, Munich, Germany
| | - Christian Kupatt
- Medical Department I, Cardiology, Angiology, Pneumology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, 81675, Munich, Germany
- DZHK (German Center of Cardiovascular Research), Munich Heart Alliance, 80336, Munich, Germany
| | - Eckhard Wolf
- Gene Center and Center for Innovative Medical Models (CiMM), LMU Munich, 81377, Munich, Germany.
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377, Munich, Germany.
| |
Collapse
|
6
|
Baine S, Wier C, Lemmerman L, Cooper-Olson G, Kempton A, Haile A, Endres J, Fedoce A, Nesbit E, Rodino-Klapac LR, Potter RA. Long-Term Survival and Myocardial Function Following Systemic Delivery of Delandistrogene Moxeparvovec in DMD MDX Rats. Hum Gene Ther 2024; 35:978-988. [PMID: 39607794 DOI: 10.1089/hum.2024.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Delandistrogene moxeparvovec is a gene transfer therapy for Duchenne muscular dystrophy (DMD) that uses an adeno-associated viral vector to deliver a micro-dystrophin transgene to skeletal and cardiac muscle. This study evaluated the long-term survival and cardiac efficacy of delandistrogene moxeparvovec in a DMD-mutated (DMDMDX) rat model of DMD-related cardiomyopathy. DMDMDX male rats, aged 21-42 days, were injected with 1.33 × 1014 viral genomes/kilogram (vg/kg) delandistrogene moxeparvovec and followed for 12, 24, and 52 weeks. Ambulation was recorded via the Photobeam Activity System, whereas echocardiograms, cardiomyocyte contractility, calcium handling, and histological analysis of fibrosis were used to evaluate cardiac disease at 12-, 24-, and 52-weeks post-treatment. A separate cohort of rats was used to assess the impact of delandistrogene moxeparvovec on survival. Treatment with delandistrogene moxeparvovec extended median survival in DMDMDX rats to >25 months versus the 13-month median survival in saline-control-treated DMDMDX rats. Compared with saline control, delandistrogene moxeparvovec therapy elicited statistically significant improvements across cardiac parameters approaching wild-type values with additional benefits in mobility, histopathology, and fibrosis observed. Transgene expression was maintained up to >25 months and micro-dystrophin expression was broadly distributed across skeletal and cardiac muscle. Taken together, these findings demonstrate long-term cardiac efficacy and improved survival following delandistrogene moxeparvovec treatment in DMDMDX rats.
Collapse
Affiliation(s)
- Stephen Baine
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Chris Wier
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Luke Lemmerman
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | | | - Amber Kempton
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Alex Haile
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Julian Endres
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | | | - Ellyn Nesbit
- Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | | | | |
Collapse
|
7
|
Gorji AE, Ahmadian K, Roudbari Z, Sadkowski T. Identification and analysis of differentially expressed lncRNAs and their ceRNA networks in DMD/mdx primary myoblasts. Sci Rep 2024; 14:23691. [PMID: 39390091 PMCID: PMC11467414 DOI: 10.1038/s41598-024-75221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 10/03/2024] [Indexed: 10/12/2024] Open
Abstract
This study explored the significance of long non-coding RNAs (lncRNAs), particularly their role in maintaining dystrophin protein stability and regulating myocyte proliferation and differentiation. The investigation focused on DMD/mdx mouse skeletal muscle primary myoblasts, aiming to identify lncRNAs potential as biomarkers and therapeutic targets for Duchenne muscular dystrophy (DMD). Utilizing CLC Genomics Workbench software, 554 differentially expressed lncRNAs were identified in DMD/mdx mice compared to wild-type (WT) control. Among them, 373 were upregulated, and 181 were downregulated. The study highlighted specific lncRNAs (e.g., 5930430L01Rik, Gm10143, LncRNA1490, LncRNA580) and their potential regulatory roles in DMD key genes like IGF1, FN1, TNNI1, and MYOD1. By predicting miRNA and their connections with lncRNA and mRNA (ceRNA network) using tools such as miRNet, miRSYSTEM and miRCARTA, the study revealed potential indirect regulation of Dystrophin, IGF1R and UTRN genes by identified lncRNAs (e.g. 2310001H17Rik-203, C130073E24Rik-202, LncRNA2767, 5930430L01Rik and LncRNA580). These findings suggest that the identified lncRNAs may play crucial roles in the development and progression of DMD through their regulatory influence on key gene expression, providing valuable insights for potential therapeutic interventions.
Collapse
Affiliation(s)
- Abdolvahab Ebrahimpour Gorji
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, 02-776, Poland
| | - Kasra Ahmadian
- Department Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Zahra Roudbari
- Department of Animal Science, Faculty of Agriculture, University of Jiroft, Jiroft, Iran
| | - Tomasz Sadkowski
- Department of Physiological Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, 02-776, Poland.
| |
Collapse
|
8
|
Earl CC, Javier AJ, Richards AM, Markham LW, Goergen CJ, Welc SS. Functional cardiac consequences of β-adrenergic stress-induced injury in a model of Duchenne muscular dystrophy. Dis Model Mech 2024; 17:dmm050852. [PMID: 39268580 PMCID: PMC11488649 DOI: 10.1242/dmm.050852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024] Open
Abstract
Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD); however, in the mdx mouse model of DMD, the cardiac phenotype differs from that seen in DMD-associated cardiomyopathy. Although some have used pharmacologic stress to stimulate injury and enhance cardiac pathology in the mdx model, many methods lead to high mortality with variable cardiac outcomes, and do not recapitulate the structural and functional cardiac changes seen in human disease. Here, we describe a simple and effective method to enhance the cardiac phenotype model in mdx mice using advanced 2D and 4D high-frequency ultrasound to monitor cardiac dysfunction progression in vivo. mdx and wild-type mice received daily low-dose (2 mg/kg/day) isoproterenol injections for 10 days. Histopathological assessment showed that isoproterenol treatment increased myocyte injury, elevated serum cardiac troponin I levels and enhanced fibrosis in mdx mice. Ultrasound revealed reduced ventricular function, decreased wall thickness, increased volumes and diminished cardiac reserve in mdx compared to wild-type mice. Our findings highlight the utility of challenging mdx mice with low-dose isoproterenol as a valuable model for exploring therapies targeting DMD-associated cardiac pathologies.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/complications
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/physiopathology
- Mice, Inbred mdx
- Isoproterenol/pharmacology
- Disease Models, Animal
- Fibrosis
- Stress, Physiological/drug effects
- Receptors, Adrenergic, beta/metabolism
- Myocardium/pathology
- Myocardium/metabolism
- Heart/drug effects
- Heart/physiopathology
- Mice
- Male
- Mice, Inbred C57BL
- Troponin I/metabolism
- Troponin I/blood
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/metabolism
- Adrenergic beta-Agonists/pharmacology
Collapse
Affiliation(s)
- Conner C. Earl
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Medicine, Indiana University School of Medicine, IN 46202, USA
| | - Areli J. Javier
- Musculoskeletal Health Sciences Program, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alyssa M. Richards
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Larry W. Markham
- Division of Pediatric Cardiology, Riley Children's Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
- Department of Medicine, Indiana University School of Medicine, IN 46202, USA
| | - Steven S. Welc
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianapolis, IN 46202, USA
| |
Collapse
|
9
|
Stirm M, Klymiuk N, Nagashima H, Kupatt C, Wolf E. Pig models for translational Duchenne muscular dystrophy research. Trends Mol Med 2024; 30:950-964. [PMID: 38749865 DOI: 10.1016/j.molmed.2024.04.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 10/12/2024]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the X-linked DMD gene, resulting in the absence of dystrophin, progressive muscle degeneration, and heart failure. Genetically tailored pig models resembling human DMD mutations recapitulate the biochemical, clinical, and pathological hallmarks of DMD with an accelerated disease progression compared to human patients. DMD pigs have been used to evaluate therapeutic concepts such as gene editing to reframe a disrupted DMD reading frame or the delivery of artificial chromosome vectors carrying the complete DMD gene. Moreover, DMD pigs have been instrumental in validating new diagnostic modalities such as multispectral optoacoustic tomography (MSOT) for non-invasive monitoring of disease progression. DMD pigs may thus help to bridge the gap between proof-of-concept studies in cellular or rodent models and clinical studies in patients.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Large Animal Models in Cardiovascular Research, Internal Medical Department I, Technical University of Munich (TU Munich), 81675 Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Kanagawa 214-8571, Japan
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum Rechts der Isar, TU Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, 81675 Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, Ludwig Maximilian University of Munich (LMU Munich), 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU, Munich, 85764, Oberschleißheim, Germany; Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
10
|
Chey YCJ, Corbett MA, Arudkumar J, Piltz SG, Thomas PQ, Adikusuma F. CRISPR-mediated megabase-scale transgene de-duplication to generate a functional single-copy full-length humanized DMD mouse model. BMC Biol 2024; 22:214. [PMID: 39334101 PMCID: PMC11438084 DOI: 10.1186/s12915-024-02008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND The development of sequence-specific precision treatments like CRISPR gene editing therapies for Duchenne muscular dystrophy (DMD) requires sequence humanized animal models to enable the direct clinical translation of tested strategies. The current available integrated transgenic mouse model containing the full-length human DMD gene, Tg(DMD)72Thoen/J (hDMDTg), has been found to have two copies of the transgene per locus in a tail-to-tail orientation, which does not accurately simulate the true (single) copy number of the DMD gene. This duplication also complicates analysis when testing CRISPR therapy editing outcomes, as large genetic alterations and rearrangements can occur between the cut sites on the two transgenes. RESULTS To address this, we performed long read nanopore sequencing on hDMDTg mice to better understand the structure of the duplicated transgenes. Following that, we performed a megabase-scale deletion of one of the transgenes by CRISPR zygotic microinjection to generate a single-copy, full-length, humanized DMD transgenic mouse model (hDMDTgSc). Functional, molecular, and histological characterisation shows that the single remaining human transgene retains its function and rescues the dystrophic phenotype caused by endogenous murine Dmd knockout. CONCLUSIONS Our unique hDMDTgSc mouse model simulates the true copy number of the DMD gene, and can potentially be used for the further generation of DMD disease models that would be better suited for the pre-clinical assessment and development of sequence specific CRISPR therapies.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Mark A Corbett
- Adelaide Medical School and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Sandra G Piltz
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- South Australian Genome Editing (SAGE) Facility, SAHMRI, Adelaide, SA, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
- South Australian Genome Editing (SAGE) Facility, SAHMRI, Adelaide, SA, Australia.
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
- Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
| |
Collapse
|
11
|
Imai S, Watanabe N, Tone Y, Mitamura R, Mori J, Kameyama T, Yamada T, Kusano K. The Usefulness of Determining Plasma and Tissue Concentrations of Phosphorodiamidate Morpholino Oligonucleotides to Estimate Their Efficacy in Duchenne Muscular Dystrophy Patients. Drug Metab Dispos 2024; 52:1029-1036. [PMID: 38991781 DOI: 10.1124/dmd.124.001806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024] Open
Abstract
Currently, four kinds of phosphorodiamidate morpholino oligomers (PMOs), such as viltolarsen, have been approved for the treatment of Duchenne muscular dystrophy (DMD); however, it is unclear whether human efficacy can be estimated using plasma concentrations. This study summarizes the tissue distribution of viltolarsen in mice and cynomolgus monkeys and evaluates the relationship between exposure and efficacy based on exon skipping. In the tissue distribution studies, all muscles in DMD-model mice showed higher concentrations of viltolarsen than those in wild-type mice and cynomolgus monkeys, and the concentrations in skeletal muscle were correlated with the exon-skipping efficiency in mice and cynomolgus monkeys. In addition, a highly sensitive bioanalytical method using liquid chromatography with tandem mass spectrometry shows promise for determining plasma concentrations up to a later time point, and the tissue (muscle)/plasma concentration ratio (Kp) in DMD-model mice was shown to be useful for predicting changes in pharmacodynamic (PD) markers in humans. Our results suggest that pharmacokinetic (PK)/PD analysis can be conducted by using the human PK profile or Kp values and skipping efficiency in DMD-model mice. This information will be useful for the efficient and effective development of PMOs as therapeutic agents. SIGNIFICANCE STATEMENT: We evaluated the relationship between the plasma or tissue concentrations and the efficiency of exon skipping for viltolarsen as an example phosphorodiamidate morpholino oligomers in the skeletal and cardiac muscle of mice and cynomolgus monkeys for pharmacokinetic/pharmacodynamic (PK/PD) analysis. The results suggest that PK/PD analysis can be conducted by using the human PK profile or tissue (muscle)/plasma concentration ratios and skipping efficiency in DMD-model mice.
Collapse
Affiliation(s)
- Shunji Imai
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Naoki Watanabe
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Yuichiro Tone
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Rei Mitamura
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Jumpei Mori
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Tsubasa Kameyama
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Tetsuhiro Yamada
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| | - Kazutomi Kusano
- Drug Metabolism and Pharmacokinetics Research Department, Discovery Research Laboratories, Kyoto (S.I., R.M., J.M., T.K., T.Y., K.K.) and Discovery Research Laboratory in Tsukuba, Ibaraki (N.W., Y.T.) , Nippon Shinyaku Co., Ltd., Japan
| |
Collapse
|
12
|
Roger AL, Biswas DD, Huston ML, Le D, Bailey AM, Pucci LA, Shi Y, Robinson-Hamm J, Gersbach CA, ElMallah MK. Respiratory characterization of a humanized Duchenne muscular dystrophy mouse model. Respir Physiol Neurobiol 2024; 326:104282. [PMID: 38782084 PMCID: PMC11472894 DOI: 10.1016/j.resp.2024.104282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/07/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024]
Abstract
Duchenne muscular dystrophy (DMD) is the most common X-linked disease. DMD is caused by a lack of dystrophin, a critical structural protein in striated muscle. Dystrophin deficiency leads to inflammation, fibrosis, and muscle atrophy. Boys with DMD have progressive muscle weakness within the diaphragm that results in respiratory failure in the 2nd or 3rd decade of life. The most common DMD mouse model - the mdx mouse - is not sufficient for evaluating genetic medicines that specifically target the human DMD (hDMD) gene sequence. Therefore, a novel transgenic mouse carrying the hDMD gene with an exon 52 deletion was created (hDMDΔ52;mdx). We characterized the respiratory function and pathology in this model using whole body plethysmography, histology, and immunohistochemistry. At 6-months-old, hDMDΔ52;mdx mice have reduced maximal respiration, neuromuscular junction pathology, and fibrosis throughout the diaphragm, which worsens at 12-months-old. In conclusion, the hDMDΔ52;mdx exhibits moderate respiratory pathology, and serves as a relevant animal model to study the impact of novel genetic therapies, including gene editing, on respiratory function.
Collapse
Affiliation(s)
- Angela L Roger
- Department of Pediatrics, Duke University, Durham, NC, USA
| | | | | | - Davina Le
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Aidan M Bailey
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Logan A Pucci
- Department of Pediatrics, Duke University, Durham, NC, USA
| | - Yihan Shi
- Department of Pediatrics, Duke University, Durham, NC, USA
| | | | | | - Mai K ElMallah
- Department of Pediatrics, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
De Masi A, Zanou N, Strotjohann K, Lee D, Lima TI, Li X, Jeon J, Place N, Jung H, Auwerx J. Cyclo His-Pro Attenuates Muscle Degeneration in Murine Myopathy Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305927. [PMID: 38728626 PMCID: PMC11267275 DOI: 10.1002/advs.202305927] [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] [Received: 08/21/2023] [Revised: 03/11/2024] [Indexed: 05/12/2024]
Abstract
Among the inherited myopathies, a group of muscular disorders characterized by structural and metabolic impairments in skeletal muscle, Duchenne muscular dystrophy (DMD) stands out for its devastating progression. DMD pathogenesis is driven by the progressive degeneration of muscle fibers, resulting in inflammation and fibrosis that ultimately affect the overall muscle biomechanics. At the opposite end of the spectrum of muscle diseases, age-related sarcopenia is a common condition that affects an increasing proportion of the elderly. Although characterized by different pathological mechanisms, DMD and sarcopenia share the development of progressive muscle weakness and tissue inflammation. Here, the therapeutic effects of Cyclo Histidine-Proline (CHP) against DMD and sarcopenia are evaluated. In the mdx mouse model of DMD, it is shown that CHP restored muscle contractility and force production, accompanied by the reduction of fibrosis and inflammation in skeletal muscle. CHP furthermore prevented the development of cardiomyopathy and fibrosis in the diaphragm, the two leading causes of death for DMD patients. CHP also attenuated muscle atrophy and functional deterioration in a mouse model of age-related sarcopenia. These findings from two different models of muscle dysfunction hence warrant further investigation into the effects of CHP on muscle pathologies in animal models and eventually in patients.
Collapse
Affiliation(s)
- Alessia De Masi
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Keno Strotjohann
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Dohyun Lee
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Tanes I. Lima
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Xiaoxu Li
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| | - Jongsu Jeon
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
| | - Nicolas Place
- Institute of Sport Sciences and Department of Biomedical SciencesFaculty of Biology‐MedicineUniversity of LausanneLausanne1015Switzerland
| | - Hoe‐Yune Jung
- R&D CenterNovMetaPharma Co., LtdPohang37668South Korea
- School of Interdisciplinary Bioscience and BioengineeringPohang University of Science and Technology (POSTECH)Pohang37673South Korea
| | - Johan Auwerx
- Laboratory of Integrative Systems PhysiologyInstitute of BioengineeringÉcole Polytechnique Fédérale de LausanneLausanne1015Switzerland
| |
Collapse
|
14
|
Soufizadeh P, Mansouri V, Ahmadbeigi N. A review of animal models utilized in preclinical studies of approved gene therapy products: trends and insights. Lab Anim Res 2024; 40:17. [PMID: 38649954 PMCID: PMC11034049 DOI: 10.1186/s42826-024-00195-6] [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: 11/09/2023] [Revised: 02/18/2024] [Accepted: 02/23/2024] [Indexed: 04/25/2024] Open
Abstract
Scientific progress heavily relies on rigorous research, adherence to scientific standards, and transparent reporting. Animal models play a crucial role in advancing biomedical research, especially in the field of gene therapy. Animal models are vital tools in preclinical research, allowing scientists to predict outcomes and understand complex biological processes. The selection of appropriate animal models is critical, considering factors such as physiological and pathophysiological similarities, availability, and ethical considerations. Animal models continue to be indispensable tools in preclinical gene therapy research. Advancements in genetic engineering and model selection have improved the fidelity and relevance of these models. As gene therapy research progresses, careful consideration of animal models and transparent reporting will contribute to the development of effective therapies for various genetic disorders and diseases. This comprehensive review explores the use of animal models in preclinical gene therapy studies for approved products up to September 2023. The study encompasses 47 approved gene therapy products, with a focus on preclinical trials. This comprehensive analysis serves as a valuable reference for researchers in the gene therapy field, aiding in the selection of suitable animal models for their preclinical investigations.
Collapse
Affiliation(s)
- Parham Soufizadeh
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Biomedical Research Institute, University of Tehran, Tehran, Iran
| | - Vahid Mansouri
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Ahmadbeigi
- Gene Therapy Research Center, Digestive Diseases Research Institute, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Earl CC, Javier AJ, Richards AM, Markham LW, Goergen CJ, Welc SS. Functional cardiac consequences of β-adrenergic stress-induced injury in the mdx mouse model of Duchenne muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589650. [PMID: 38659739 PMCID: PMC11042272 DOI: 10.1101/2024.04.15.589650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD), however, in the mdx mouse model of DMD, the cardiac phenotype differs from that seen in DMD-associated cardiomyopathy. Although some have used pharmacologic stress to enhance the cardiac phenotype in the mdx model, many methods lead to high mortality, variable cardiac outcomes, and do not recapitulate the structural and functional cardiac changes seen in human disease. Here, we describe a simple and effective method to enhance the cardiac phenotype model in mdx mice using advanced 2D and 4D high-frequency ultrasound to monitor cardiac dysfunction progression in vivo. For our study, mdx and wild-type (WT) mice received daily low-dose (2 mg/kg/day) isoproterenol injections for 10 days. Histopathologic assessment showed that isoproterenol treatment increased myocyte injury, elevated serum cardiac troponin I levels, and enhanced fibrosis in mdx mice. Ultrasound revealed reduced ventricular function, decreased wall thickness, increased volumes, and diminished cardiac reserve in mdx mice compared to wild-type. Our findings highlight the utility of low-dose isoproterenol in mdx mice as a valuable model for exploring therapies targeting DMD-associated cardiac complications.
Collapse
Affiliation(s)
- Conner C. Earl
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
- Indiana University School of Medicine, IN, USA
| | - Areli J. Javier
- Musculoskeletal Health Sciences Program, Indiana University School of Medicine, Indianapolis, IN USA
| | - Alyssa M. Richards
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
| | - Larry W. Markham
- Division of Pediatric Cardiology, Riley Children’s Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette IN, USA
- Indiana University School of Medicine, IN, USA
| | - Steven S. Welc
- Division of Pediatric Cardiology, Riley Children’s Hospital at Indiana University Health, Indiana University School of Medicine, Indianapolis, IN
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis IN, USA
| |
Collapse
|
16
|
Saxena H, Weintraub NL, Tang Y. Potential Therapeutic Targets for Hypotension in Duchenne Muscular Dystrophy. Med Hypotheses 2024; 185:111318. [PMID: 38585412 PMCID: PMC10993928 DOI: 10.1016/j.mehy.2024.111318] [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] [Indexed: 04/09/2024]
Abstract
Duchenne Muscular Dystrophy (DMD) is marked by genetic mutations occurring in the DMD gene, which is widely expressed in the cardiovascular system. In addition to developing cardiomyopathy, patients with DMD have been reported to be susceptible to the development of symptomatic hypotension, although the mechanisms are unclear. Analysis of single-cell RNA sequencing data has identified potassium voltage-gated channel subfamily Q member 5 (KCNQ5) and possibly ryanodine receptor 2 (RyR2) as potential candidate hypotension genes whose expression is significantly upregulated in the vascular smooth muscle cells of DMD mutant mice. We hypothesize that heightened KCNQ5 and RyR2 expression contributes to decreased arterial blood pressure in patients with DMD. Exploring pharmacological approaches to inhibit the KCNQ5 and RyR2 channels holds promise in managing the systemic hypotension observed in individuals with DMD. This avenue of investigation presents new prospects for improving clinical outcomes for these patients.
Collapse
Affiliation(s)
- Harshi Saxena
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| | - Yaoliang Tang
- Vascular Biology Center, Department of Medicine, Medical College of Georgia at Augusta University, 1460 Laney Walker Blvd, Augusta, GA 30912, USA
| |
Collapse
|
17
|
Day KS, Rempel L, Rossi FMV, Theret M. Origins and functions of eosinophils in two non-mucosal tissues. Front Immunol 2024; 15:1368142. [PMID: 38585275 PMCID: PMC10995313 DOI: 10.3389/fimmu.2024.1368142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
Eosinophils are a type of granulocyte named after the presence of their eosin-stained granules. Traditionally, eosinophils have been best known to play prominent roles in anti-parasitic responses and mediating allergic reactions. Knowledge of their behaviour has expanded with time, and they are now recognized to play integral parts in the homeostasis of gastrointestinal, respiratory, skeletal muscle, adipose, and connective tissue systems. As such, they are implicated in a myriad of pathologies, and have been the target of several medical therapies. This review focuses on the lifespan of eosinophils, from their origins in the bone marrow, to their tissue-resident role. In particular, we wish to highlight the functions of eosinophils in non-mucosal tissues with skeletal muscle and the adipose tissues as examples, and to discuss the current understanding of their participation in diseased states in these tissues.
Collapse
Affiliation(s)
- Katie S. Day
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Lucas Rempel
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M. V. Rossi
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Marine Theret
- Department of Medical Genetics, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Timpani CA, Kourakis S, Debruin DA, Campelj DG, Pompeani N, Dargahi N, Bautista AP, Bagaric RM, Ritenis EJ, Sahakian L, Debrincat D, Stupka N, Hafner P, Arthur PG, Terrill JR, Apostolopoulos V, de Haan JB, Guven N, Fischer D, Rybalka E. Dimethyl fumarate modulates the dystrophic disease program following short-term treatment. JCI Insight 2023; 8:e165974. [PMID: 37751291 PMCID: PMC10721277 DOI: 10.1172/jci.insight.165974] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 09/20/2023] [Indexed: 09/27/2023] Open
Abstract
New medicines are urgently required to treat the fatal neuromuscular disease Duchenne muscular dystrophy (DMD). Dimethyl fumarate (DMF) is a potent immunomodulatory small molecule nuclear erythroid 2-related factor 2 activator with current clinical utility in the treatment of multiple sclerosis and psoriasis that could be effective for DMD and rapidly translatable. Here, we tested 2 weeks of daily 100 mg/kg DMF versus 5 mg/kg standard-care prednisone (PRED) treatment in juvenile mdx mice with early symptomatic DMD. Both drugs modulated seed genes driving the DMD disease program and improved force production in fast-twitch muscle. However, only DMF showed pro-mitochondrial effects, protected contracting muscles from fatigue, improved histopathology, and augmented clinically compatible muscle function tests. DMF may be a more selective modulator of the DMD disease program than PRED, warranting follow-up longitudinal studies to evaluate disease-modifying impact.
Collapse
Affiliation(s)
- Cara A. Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine – Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Stephanie Kourakis
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Danielle A. Debruin
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Dean G. Campelj
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
| | - Nancy Pompeani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Florey Institute of Neuroscience and Mental Health, Heidelberg, Victoria, Australia
| | - Narges Dargahi
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
| | - Angelo P. Bautista
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Ryan M. Bagaric
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Elya J. Ritenis
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Lauren Sahakian
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Department of Medicine – Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Didier Debrincat
- College of Health and Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Nicole Stupka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine – Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Patricia Hafner
- Division of Neuropaediatrics and Developmental Medicine, University Children’s Hospital of Basel (UKBB), Basel, Switzerland
| | - Peter G. Arthur
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Jessica R. Terrill
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
| | - Judy B. de Haan
- Basic Science Domain, Oxidative Stress Laboratory, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, Victoria, Australia
- Faculty of Science, Engineering and Technology, Swinburne University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Parkville, Victoria, Australia
| | - Nuri Guven
- Department of Medicine – Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
| | - Dirk Fischer
- Division of Neuropaediatrics and Developmental Medicine, University Children’s Hospital of Basel (UKBB), Basel, Switzerland
| | - Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), St Albans, Victoria, Australia
- Department of Medicine – Western Health, Melbourne Medical School, The University of Melbourne, St Albans, Victoria, Australia
- Division of Neuropaediatrics and Developmental Medicine, University Children’s Hospital of Basel (UKBB), Basel, Switzerland
| |
Collapse
|
19
|
Lee J, Myrie NO, Jeong GJ, Han WM, Jang YC, García AJ, Emelianov S. In vivo shear wave elasticity imaging for assessment of diaphragm function in muscular dystrophy. Acta Biomater 2023; 168:277-285. [PMID: 37453552 PMCID: PMC10540053 DOI: 10.1016/j.actbio.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/28/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Duchenne muscular dystrophy (DMD) causes patients to suffer from ambulatory disability and cardiorespiratory failure, the latter of which leads to premature death. Due to its role in respiration, the diaphragm is an important muscle for study. A common method for evaluating diaphragm function is ex vivo force testing, which only allows for an end point measurement. In contrast, ultrasound shear wave elastography imaging (US-SWEI) can assess diaphragm function over time; however, US-SWEI studies in dystrophic patients to date have focused on the limbs without preclinical studies. In this work, we used US-SWEI to estimate the shear wave speed (SWS) in diaphragm muscles of healthy (WT) mice, mdx mice, and mdx mice haploinsufficient for utrophin (mdx-utr) at 6 and 12 months of age. Diaphragms were then subjected to ex vivo force testing and histological analysis at 12 months of age. Between 6 and 12 months, a 23.8% increase in SWS was observed in WT mice and a 27.8% increase in mdx mice, although no significant difference was found in mdx-utr mice. Specific force generated by mdx-utr diaphragms was lower than that of WT diaphragms following twitch stimulus. A strong correlation between SWS and collagen deposition was observed, as well as between SWS and muscle fiber size. Together, these data demonstrate the ability of US-SWEI to evaluate dystrophic diaphragm functionality over time and predict the biochemical and morphological make-up of the diaphragm. Additionally, our results highlight the advantage of US-SWEI over ex vivo testing by obtaining longitudinal measurements in the same subject. STATEMENT OF SIGNIFICANCE: In DMD patients, muscles experience cycles of regeneration and degeneration that contribute to chronic inflammation and muscle weakness. This pathology only worsens with time and leads to muscle wasting, including in respiratory and cardiac muscles. Because respiratory failure is a major contributor to premature death in DMD patients, the diaphragm muscle is an important muscle to evaluate and treat over time. Currently, diaphragm function is assessed using ex vivo force testing, a technique that only allows measurement at sacrifice. In contrast, ultrasonography, particularly shear wave elasticity imaging (USSWEI), is a promising tool for longitudinal assessment; however, most US-SWEI in DMD patients aimed for limb muscles only with the absence of preclinical studies. This work broadens the applications of US-SWE imaging by demonstrating its ability to track properties and function of dystrophic diaphragm muscles longitudinally in multiple dystrophic mouse models.
Collapse
Affiliation(s)
- Jeehyun Lee
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Nia O Myrie
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Gun-Jae Jeong
- Institute of Cell and Tissue Engineering, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Woojin M Han
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Young C Jang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA; Department of Orthopedics, Emory Musculoskeletal Institute, Emory School of Medicine, Atlanta, GA 30329, USA.
| | - Andrés J García
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Stanislav Emelianov
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA.
| |
Collapse
|
20
|
Dowling P, Gargan S, Zweyer M, Swandulla D, Ohlendieck K. Extracellular Matrix Proteomics: The mdx-4cv Mouse Diaphragm as a Surrogate for Studying Myofibrosis in Dystrophinopathy. Biomolecules 2023; 13:1108. [PMID: 37509144 PMCID: PMC10377647 DOI: 10.3390/biom13071108] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
The progressive degeneration of the skeletal musculature in Duchenne muscular dystrophy is accompanied by reactive myofibrosis, fat substitution, and chronic inflammation. Fibrotic changes and reduced tissue elasticity correlate with the loss in motor function in this X-chromosomal disorder. Thus, although dystrophinopathies are due to primary abnormalities in the DMD gene causing the almost-complete absence of the cytoskeletal Dp427-M isoform of dystrophin in voluntary muscles, the excessive accumulation of extracellular matrix proteins presents a key histopathological hallmark of muscular dystrophy. Animal model research has been instrumental in the characterization of dystrophic muscles and has contributed to a better understanding of the complex pathogenesis of dystrophinopathies, the discovery of new disease biomarkers, and the testing of novel therapeutic strategies. In this article, we review how mass-spectrometry-based proteomics can be used to study changes in key components of the endomysium, perimysium, and epimysium, such as collagens, proteoglycans, matricellular proteins, and adhesion receptors. The mdx-4cv mouse diaphragm displays severe myofibrosis, making it an ideal model system for large-scale surveys of systematic alterations in the matrisome of dystrophic fibers. Novel biomarkers of myofibrosis can now be tested for their appropriateness in the preclinical and clinical setting as diagnostic, pharmacodynamic, prognostic, and/or therapeutic monitoring indicators.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Stephen Gargan
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
| | - Margit Zweyer
- Department of Neonatology and Paediatric Intensive Care, Children's Hospital, German Center for Neurodegenerative Diseases, University of Bonn, D53127 Bonn, Germany
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, D53115 Bonn, Germany
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, National University of Ireland, W23 F2H6 Maynooth, Co. Kildare, Ireland
| |
Collapse
|
21
|
George TG, Hanft LM, Krenz M, Domeier TL, McDonald KS. Dystrophic cardiomyopathy: role of the cardiac myofilaments. Front Physiol 2023; 14:1207658. [PMID: 37362434 PMCID: PMC10288979 DOI: 10.3389/fphys.2023.1207658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
Dystrophic cardiomyopathy arises from mutations in the dystrophin gene. Dystrophin forms part of the dystrophin glycoprotein complex and is postulated to act as a membrane stabilizer, protecting the sarcolemma from contraction-induced damage. Duchenne muscular dystrophy (DMD) is the most severe dystrophinopathy, caused by a total absence of dystrophin. Patients with DMD present with progressive skeletal muscle weakness and, because of treatment advances, a cardiac component of the disease (i.e., dystrophic cardiomyopathy) has been unmasked later in disease progression. The role that myofilaments play in dystrophic cardiomyopathy is largely unknown and, as such, this study aimed to address cardiac myofilament function in a mouse model of muscular dystrophy. To assess the effects of DMD on myofilament function, isolated permeabilized cardiomyocytes of wild-type (WT) littermates and Dmdmdx-4cv mice were attached between a force transducer and motor and subjected to contractile assays. Maximal tension and rates of force development (indexed by the rate constant, k tr) were similar between WT and Dmdmdx-4cv cardiac myocyte preparations. Interestingly, Dmdmdx-4cv cardiac myocytes exhibited greater sarcomere length dependence of peak power output compared to WT myocyte preparations. These results suggest dystrophin mitigates length dependence of activation and, in the absence of dystrophin, augmented sarcomere length dependence of myocyte contractility may accelerate ventricular myocyte contraction-induced damage and contribute to dystrophic cardiomyopathy. Next, we assessed if mavacamten, a small molecule modulator of thick filament activation, would mitigate contractile properties observed in Dmdmdx-4cv permeabilized cardiac myocyte preparations. Mavacamten decreased maximal tension and k tr in both WT and Dmdmdx-4cv cardiac myocytes, while also normalizing the length dependence of peak power between WT and Dmdmdx-4cv cardiac myocyte preparations. These results highlight potential benefits of mavacamten (i.e., reduced contractility while maintaining exquisite sarcomere length dependence of power output) as a treatment for dystrophic cardiomyopathy associated with DMD.
Collapse
|
22
|
Acin‐Perez R, Benincá C, Fernandez del Rio L, Shu C, Baghdasarian S, Zanette V, Gerle C, Jiko C, Khairallah R, Khan S, Rincon Fernandez Pacheco D, Shabane B, Erion K, Masand R, Dugar S, Ghenoiu C, Schreiner G, Stiles L, Liesa M, Shirihai OS. Inhibition of ATP synthase reverse activity restores energy homeostasis in mitochondrial pathologies. EMBO J 2023; 42:e111699. [PMID: 36912136 PMCID: PMC10183817 DOI: 10.15252/embj.2022111699] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 03/14/2023] Open
Abstract
The maintenance of cellular function relies on the close regulation of adenosine triphosphate (ATP) synthesis and hydrolysis. ATP hydrolysis by mitochondrial ATP Synthase (CV) is induced by loss of proton motive force and inhibited by the mitochondrial protein ATPase inhibitor (ATPIF1). The extent of CV hydrolytic activity and its impact on cellular energetics remains unknown due to the lack of selective hydrolysis inhibitors of CV. We find that CV hydrolytic activity takes place in coupled intact mitochondria and is increased by respiratory chain defects. We identified (+)-Epicatechin as a selective inhibitor of ATP hydrolysis that binds CV while preventing the binding of ATPIF1. In cells with Complex-III deficiency, we show that inhibition of CV hydrolytic activity by (+)-Epichatechin is sufficient to restore ATP content without restoring respiratory function. Inhibition of CV-ATP hydrolysis in a mouse model of Duchenne Muscular Dystrophy is sufficient to improve muscle force without any increase in mitochondrial content. We conclude that the impact of compromised mitochondrial respiration can be lessened using hydrolysis-selective inhibitors of CV.
Collapse
Affiliation(s)
- Rebeca Acin‐Perez
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cristiane Benincá
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Lucia Fernandez del Rio
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Cynthia Shu
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Siyouneh Baghdasarian
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | - Vanessa Zanette
- Department of BioinformaticsUniversity Federal of ParanaCuritibaBrazil
| | - Christoph Gerle
- Institute for Protein ResearchOsaka UniversitySuitaJapan
- RIKEN SPring‐8 CenterSayo‐gunJapan
| | - Chimari Jiko
- Institute for Integrated Radiation and Nuclear ScienceKyoto UniversityKyotoJapan
| | | | | | | | - Byourak Shabane
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
| | | | | | | | | | | | - Linsey Stiles
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Marc Liesa
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
- Institut de Biologia Molecular de Barcelona, IBMB, CSICBarcelonaCataloniaSpain
| | - Orian S Shirihai
- Department of Medicine, Endocrinology, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Metabolism Theme, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCAUSA
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Molecular Cellular Integrative PhysiologyUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
23
|
Bobadilla Muñoz M, Orbe J, Abizanda G, Machado FJD, Vilas A, Ullate-Agote A, Extramiana L, Baraibar Churio A, Aranguren XL, Cantero G, Sáinz Amillo N, Rodríguez JA, Ramos García L, Romero Riojas JP, Vallejo-Illarramendi A, Paradas C, López de Munain A, Páramo JA, Prósper F, Pérez-Ruiz A. Loss of the matrix metalloproteinase-10 causes premature features of aging in satellite cells. Front Cell Dev Biol 2023; 11:1128534. [PMID: 37228645 PMCID: PMC10203875 DOI: 10.3389/fcell.2023.1128534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
Aged muscles accumulate satellite cells with a striking decline response to damage. Although intrinsic defects in satellite cells themselves are the major contributors to aging-associated stem cell dysfunction, increasing evidence suggests that changes in the muscle-stem cell local microenvironment also contribute to aging. Here, we demonstrate that loss of the matrix metalloproteinase-10 (MMP-10) in young mice alters the composition of the muscle extracellular matrix (ECM), and specifically disrupts the extracellular matrix of the satellite cell niche. This situation causes premature features of aging in the satellite cells, contributing to their functional decline and a predisposition to enter senescence under proliferative pressure. Similarly, reduction of MMP-10 levels in young satellite cells from wild type animals induces a senescence response, while addition of the protease delays this program. Significantly, the effect of MMP-10 on satellite cell aging can be extended to another context of muscle wasting, muscular dystrophy. Systemic treatment of mdx dystrophic mice with MMP-10 prevents the muscle deterioration phenotype and reduces cellular damage in the satellite cells, which are normally under replicative pressure. Most importantly, MMP-10 conserves its protective effect in the satellite cell-derived myoblasts isolated from a Duchenne muscular dystrophy patient by decreasing the accumulation of damaged DNA. Hence, MMP-10 provides a previously unrecognized therapeutic opportunity to delay satellite cell aging and overcome satellite cell dysfunction in dystrophic muscles.
Collapse
Affiliation(s)
- Miriam Bobadilla Muñoz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Josune Orbe
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS)-Ictus, Instituto de Salud Carlos III, Madrid, Spain
| | - Gloria Abizanda
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Florencio J. D. Machado
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Amaia Vilas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Asier Ullate-Agote
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Leire Extramiana
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Arantxa Baraibar Churio
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Xabier L. Aranguren
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gloria Cantero
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Neira Sáinz Amillo
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Centre for Nutrition Research, Universidad de Navarra, Pamplona, Spain
| | - José Antonio Rodríguez
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Luis Ramos García
- Radiology Department, Clínica Universidad de Navarra, Pamplona, Spain
- Radiology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - Juan Pablo Romero Riojas
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | | | - Carmen Paradas
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Neuromuscular Disorders Unit, Sevilla, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Adolfo López de Munain
- CIBERNED-Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
- Neurology Department, Osakidetza Basque Health Service, Donostialdea Integrated Health Organisation, San Sebastian, Spain
| | - José Antonio Páramo
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
- Centro de Investigación en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prósper
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Pamplona, Spain
| | - Ana Pérez-Ruiz
- Regenerative Medicine Program, Center for Applied Medical Research (CIMA) Universidad de Navarra, CIBERONC, Madrid, Spain
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| |
Collapse
|
24
|
Krishna S, Spaulding HR, Koltes JE, Quindry JC, Valentine RJ, Selsby JT. Indicators of increased ER stress and UPR in aged D2-mdx and human dystrophic skeletal muscles. Front Physiol 2023; 14:1152576. [PMID: 37179835 PMCID: PMC10166835 DOI: 10.3389/fphys.2023.1152576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/10/2023] [Indexed: 05/15/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease that results in muscle wasting, wheelchair dependence, and eventual death due to cardiac and respiratory complications. In addition to muscle fragility, dystrophin deficiency also results in multiple secondary dysfunctions, which may lead to the accumulation of unfolded proteins causing endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). The purpose of this investigation was to understand how ER stress and the UPR are modified in muscle from D2-mdx mice, an emerging DMD model, and from humans with DMD. We hypothesized that markers of ER stress and the UPR are upregulated in D2-mdx and human dystrophic muscles compared to their healthy counterparts. Immunoblotting in diaphragms from 11-month-old D2-mdx and DBA mice indicated increased ER stress and UPR in dystrophic diaphragms compared to healthy, including increased relative abundance of ER stress chaperone CHOP, canonical ER stress transducers ATF6 and pIRE1α S724, and transcription factors that regulate the UPR such as ATF4, XBP1s, and peIF2α S51. The publicly available Affymetrix dataset (GSE38417) was used to analyze the expression of ER stress and UPR-related transcripts and processes. Fifty-eight upregulated genes related to ER stress and the UPR in human dystrophic muscles suggest pathway activation. Further, based on analyses using iRegulon, putative transcription factors that regulate this upregulation profile were identified, including ATF6, XBP1, ATF4, CREB3L2, and EIF2AK3. This study adds to and extends the emerging knowledge of ER stress and the UPR in dystrophin deficiency and identifies transcriptional regulators that may be responsible for these changes and be of therapeutic interest.
Collapse
Affiliation(s)
- Swathy Krishna
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - Hannah R. Spaulding
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - James E. Koltes
- Department of Animal Science, Iowa State University, Ames, IA, United States
| | - John C. Quindry
- School of Integrative Physiology and Athletic Training, University of Montana, Missoula, MT, United States
| | - Rudy J. Valentine
- Department of Kinesiology, Iowa State University, Ames, IA, United States
| | - Joshua T. Selsby
- Department of Animal Science, Iowa State University, Ames, IA, United States
| |
Collapse
|
25
|
Tang JM, McClennan A, Liu L, Hadway J, Ronald JA, Hicks JW, Hoffman L, Anazodo UC. A Protocol for Simultaneous In Vivo Imaging of Cardiac and Neuroinflammation in Dystrophin-Deficient MDX Mice Using [ 18F]FEPPA PET. Int J Mol Sci 2023; 24:ijms24087522. [PMID: 37108685 PMCID: PMC10144317 DOI: 10.3390/ijms24087522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder caused by dystrophin loss-notably within muscles and the central neurons system. DMD presents as cognitive weakness, progressive skeletal and cardiac muscle degeneration until pre-mature death from cardiac or respiratory failure. Innovative therapies have improved life expectancy; however, this is accompanied by increased late-onset heart failure and emergent cognitive degeneration. Thus, better assessment of dystrophic heart and brain pathophysiology is needed. Chronic inflammation is strongly associated with skeletal and cardiac muscle degeneration; however, neuroinflammation's role is largely unknown in DMD despite being prevalent in other neurodegenerative diseases. Here, we present an inflammatory marker translocator protein (TSPO) positron emission tomography (PET) protocol for in vivo concomitant assessment of immune cell response in hearts and brains of a dystrophin-deficient mouse model [mdx:utrn(+/-)]. Preliminary analysis of whole-body PET imaging using the TSPO radiotracer, [18F]FEPPA in four mdx:utrn(+/-) and six wildtype mice are presented with ex vivo TSPO-immunofluorescence tissue staining. The mdx:utrn(+/-) mice showed significant elevations in heart and brain [18F]FEPPA activity, which correlated with increased ex vivo fluorescence intensity, highlighting the potential of TSPO-PET to simultaneously assess presence of cardiac and neuroinflammation in dystrophic heart and brain, as well as in several organs within a DMD model.
Collapse
Affiliation(s)
- Joanne M Tang
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Andrew McClennan
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Linshan Liu
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Jennifer Hadway
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - John A Ronald
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Robarts Research Institute, Western University, London, ON N6A 3K7, Canada
| | - Justin W Hicks
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Lisa Hoffman
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
- Department of Anatomy and Cell Biology, Western University, London, ON N6A 3K7, Canada
| | - Udunna C Anazodo
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
26
|
Abbas H, Olivere LA, Padgett ME, Schmidt CA, Gilmore BF, McCord TJ, Southerland KW, McClung JM, Kontos CD. Muscle progenitor cells are required for skeletal muscle regeneration and prevention of adipogenesis after limb ischemia. Front Cardiovasc Med 2023; 10:1118738. [PMID: 36937923 PMCID: PMC10017542 DOI: 10.3389/fcvm.2023.1118738] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/08/2023] [Indexed: 03/06/2023] Open
Abstract
Skeletal muscle injury in peripheral artery disease (PAD) has been attributed to vascular insufficiency, however evidence has demonstrated that muscle cell responses play a role in determining outcomes in limb ischemia. Here, we demonstrate that genetic ablation of Pax7+ muscle progenitor cells (MPCs) in a model of hindlimb ischemia (HLI) inhibited muscle regeneration following ischemic injury, despite a lack of morphological or physiological changes in resting muscle. Compared to control mice (Pax7WT), the ischemic limb of Pax7-deficient mice (Pax7Δ) was unable to generate significant force 7 or 28 days after HLI. A significant increase in adipose was observed in the ischemic limb 28 days after HLI in Pax7Δ mice, which replaced functional muscle. Adipogenesis in Pax7Δ mice corresponded with a significant increase in PDGFRα+ fibro/adipogenic progenitors (FAPs). Inhibition of FAPs with batimastat decreased muscle adipose but increased fibrosis. In vitro, Pax7Δ MPCs failed to form myotubes but displayed increased adipogenesis. Skeletal muscle from patients with critical limb threatening ischemia displayed increased adipose in more ischemic regions of muscle, which corresponded with fewer satellite cells. Collectively, these data demonstrate that Pax7+ MPCs are required for muscle regeneration after ischemia and suggest that muscle regeneration may be an important therapeutic target in PAD.
Collapse
Affiliation(s)
- Hasan Abbas
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | | | - Michael E. Padgett
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
| | - Cameron A. Schmidt
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
| | - Brian F. Gilmore
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Timothy J. McCord
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Kevin W. Southerland
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Joseph M. McClung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Brody School of Medicine, East Carolina Heart Institute, East Carolina University, Greenville, NC, United States
| | - Christopher D. Kontos
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, United States
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, United States
- Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
27
|
Sarcoplasmic Reticulum Ca 2+ Buffer Proteins: A Focus on the Yet-To-Be-Explored Role of Sarcalumenin in Skeletal Muscle Health and Disease. Cells 2023; 12:cells12050715. [PMID: 36899851 PMCID: PMC10000884 DOI: 10.3390/cells12050715] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/06/2023] Open
Abstract
Sarcalumenin (SAR) is a luminal Ca2+ buffer protein with high capacity but low affinity for calcium binding found predominantly in the longitudinal sarcoplasmic reticulum (SR) of fast- and slow-twitch skeletal muscles and the heart. Together with other luminal Ca2+ buffer proteins, SAR plays a critical role in modulation of Ca2+ uptake and Ca2+ release during excitation-contraction coupling in muscle fibers. SAR appears to be important in a wide range of other physiological functions, such as Sarco-Endoplasmic Reticulum Calcium ATPase (SERCA) stabilization, Store-Operated-Calcium-Entry (SOCE) mechanisms, muscle fatigue resistance and muscle development. The function and structural features of SAR are very similar to those of calsequestrin (CSQ), the most abundant and well-characterized Ca2+ buffer protein of junctional SR. Despite the structural and functional similarity, very few targeted studies are available in the literature. The present review provides an overview of the role of SAR in skeletal muscle physiology, as well as of its possible involvement and dysfunction in muscle wasting disorders, in order to summarize the current knowledge on SAR and drive attention to this important but still underinvestigated/neglected protein.
Collapse
|
28
|
Chey YCJ, Arudkumar J, Aartsma-Rus A, Adikusuma F, Thomas PQ. CRISPR applications for Duchenne muscular dystrophy: From animal models to potential therapies. WIREs Mech Dis 2023; 15:e1580. [PMID: 35909075 PMCID: PMC10078488 DOI: 10.1002/wsbm.1580] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/28/2022] [Accepted: 06/30/2022] [Indexed: 01/31/2023]
Abstract
CRISPR gene-editing technology creates precise and permanent modifications to DNA. It has significantly advanced our ability to generate animal disease models for use in biomedical research and also has potential to revolutionize the treatment of genetic disorders. Duchenne muscular dystrophy (DMD) is a monogenic muscle-wasting disease that could potentially benefit from the development of CRISPR therapy. It is commonly associated with mutations that disrupt the reading frame of the DMD gene that encodes dystrophin, an essential scaffolding protein that stabilizes striated muscles and protects them from contractile-induced damage. CRISPR enables the rapid generation of various animal models harboring mutations that closely simulates the wide variety of mutations observed in DMD patients. These models provide a platform for the testing of sequence-specific interventions like CRISPR therapy that aim to reframe or skip DMD mutations to restore functional dystrophin expression. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics.
Collapse
Affiliation(s)
- Yu C J Chey
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jayshen Arudkumar
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fatwa Adikusuma
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,CSIRO Synthetic Biology Future Science Platform, Canberra, Australia
| | - Paul Q Thomas
- School of Biomedicine and Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.,Genome Editing Program, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia.,South Australian Genome Editing (SAGE), South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| |
Collapse
|
29
|
Donen G, Milad N, Bernatchez P. Humanization of the mdx Mouse Phenotype for Duchenne Muscular Dystrophy Modeling: A Metabolic Perspective. J Neuromuscul Dis 2023; 10:1003-1012. [PMID: 37574742 PMCID: PMC10657711 DOI: 10.3233/jnd-230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/15/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe form of muscular dystrophy (MD) that is characterized by early muscle wasting and lethal cardiorespiratory failure. While the mdx mouse is the most common model of DMD, it fails to replicate the severe loss of muscle mass and other complications observed in patients, in part due to the multiple rescue pathways found in mice. This led to several attempts at improving DMD animal models by interfering with these rescue pathways through double transgenic approaches, resulting in more severe phenotypes with mixed relevance to the human pathology. As a growing body of literature depicts DMD as a multi-system metabolic disease, improvements in mdx-based modeling of DMD may be achieved by modulating whole-body metabolism instead of muscle homeostasis. This review provides an overview of the established dual-transgenic approaches that exacerbate the mild mdx phenotype by primarily interfering with muscle homeostasis and highlights how advances in DMD modeling coincide with inducing whole-body metabolic changes. We focus on the DBA2/J strain-based D2.mdx mouse with heightened transforming growth factor (TGF)-β signaling and the dyslipidemic mdx/apolipoprotein E (mdx/ApoE) knock-out (KO) mouse, and summarize how these novel models emulate the metabolic changes observed in DMD.
Collapse
Affiliation(s)
| | | | - Pascal Bernatchez
- Correspondence to: Dr. Pascal Bernatchez, Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, 2176 Health Sciences mall, room 217, Vancouver BC, V6T 1Z3, Canada. Tel.: +1 604 806 8346 /Ext.66060; E-mail:
| |
Collapse
|
30
|
Dubuisson N, Versele R, Planchon C, Selvais CM, Noel L, Abou-Samra M, Davis-López de Carrizosa MA. Histological Methods to Assess Skeletal Muscle Degeneration and Regeneration in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:16080. [PMID: 36555721 PMCID: PMC9786356 DOI: 10.3390/ijms232416080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease caused by the loss of function of the protein dystrophin. This protein contributes to the stabilisation of striated cells during contraction, as it anchors the cytoskeleton with components of the extracellular matrix through the dystrophin-associated protein complex (DAPC). Moreover, absence of the functional protein affects the expression and function of proteins within the DAPC, leading to molecular events responsible for myofibre damage, muscle weakening, disability and, eventually, premature death. Presently, there is no cure for DMD, but different treatments help manage some of the symptoms. Advances in genetic and exon-skipping therapies are the most promising intervention, the safety and efficiency of which are tested in animal models. In addition to in vivo functional tests, ex vivo molecular evaluation aids assess to what extent the therapy has contributed to the regenerative process. In this regard, the later advances in microscopy and image acquisition systems and the current expansion of antibodies for immunohistological evaluation together with the development of different spectrum fluorescent dyes have made histology a crucial tool. Nevertheless, the complexity of the molecular events that take place in dystrophic muscles, together with the rise of a multitude of markers for each of the phases of the process, makes the histological assessment a challenging task. Therefore, here, we summarise and explain the rationale behind different histological techniques used in the literature to assess degeneration and regeneration in the field of dystrophinopathies, focusing especially on those related to DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Neuromuscular Reference Center, Cliniques Universitaires Saint-Luc (CUSL), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
31
|
Choi MC, Jo J, Lee M, Park J, Yao TP, Park Y. Cathelicidin-related antimicrobial peptide mediates skeletal muscle degeneration caused by injury and Duchenne muscular dystrophy in mice. J Cachexia Sarcopenia Muscle 2022; 13:3091-3105. [PMID: 36059045 PMCID: PMC9745559 DOI: 10.1002/jcsm.13065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cathelicidin, an antimicrobial peptide, plays a key role in regulating bacterial killing and innate immunity; however, its role in skeletal muscle function is unknown. We investigated the potential role of cathelicidin in skeletal muscle pathology resulting from acute injury and Duchenne muscular dystrophy (DMD) in mice. METHODS Expression changes and muscular localization of mouse cathelicidin-related antimicrobial peptide (Cramp) were examined in the skeletal muscle of normal mice treated with chemicals (cardiotoxin and BaCl2 ) or in dystrophic muscle of DMD mouse models (mdx, mdx/Utrn+/- and mdx/Utrn-/- ). Cramp penetration into myofibres and effects on muscle damage were studied by treating synthetic peptides to mouse skeletal muscles or C2C12 myotubes. Cramp knockout (KO) mice and mdx/Utrn/Cramp KO lines were used to determine whether Cramp mediates muscle degeneration. Muscle pathophysiology was assessed by histological methods, serum analysis, grip strength and lifespan. Molecular factors targeted by Cramp were identified by the pull-down assay and proteomic analysis. RESULTS In response to acute muscle injury, Cramp was activated in muscle-infiltrating neutrophils and internalized into myofibres. Cramp treatments of mouse skeletal muscles or C2C12 myotubes resulted in muscle degeneration and myotube damage, respectively. Genetic ablation of Cramp reduced neutrophil infiltration and ameliorated muscle pathology, such as fibre size (P < 0.001; n = 6) and fibrofatty infiltration (P < 0.05). Genetic reduction of Cramp in mdx/Utrn+/- mice not only attenuated muscle damage (35%, P < 0.05; n = 9-10), myonecrosis (53%, P < 0.05), inflammation (37-65%, P < 0.01) and fibrosis (14%, P < 0.05) but also restored muscle fibre size (14%, P < 0.05) and muscle force (18%, P < 0.05). Reducing Cramp levels led to a 63% (male, P < 0.05; n = 10-14) and a 124% (female, P < 0.001; n = 20) increase in the lifespan of mdx/Utrn-/- mice. Proteomic and mechanistic studies revealed that Cramp cross-talks with Ca2+ signalling in skeletal muscle through sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase1 (SERCA1). Cramp binds and inactivates SERCA1, leading to the activation of Ca2+ -dependent calpain proteases that exacerbate DMD progression. CONCLUSIONS These findings identify Cramp as an immune cell-derived regulator of skeletal muscle degeneration and provide a potential therapeutic target for DMD.
Collapse
Affiliation(s)
- Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jiwon Jo
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Myeongjin Lee
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jonggwan Park
- Department of Bioinformatics, Kongju National University, Kongju, South Korea
| | - Tso-Pang Yao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| |
Collapse
|
32
|
Himelman E, Nouet J, Lillo MA, Chong A, Zhou D, Wehrens XHT, Rodney GG, Xie LH, Shirokova N, Contreras JE, Fraidenraich D. A microtubule-connexin-43 regulatory link suppresses arrhythmias and cardiac fibrosis in Duchenne muscular dystrophy mice. Am J Physiol Heart Circ Physiol 2022; 323:H983-H995. [PMID: 36206047 PMCID: PMC9639757 DOI: 10.1152/ajpheart.00179.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022]
Abstract
Dilated cardiomyopathy is the leading cause of death in Duchenne muscular dystrophy (DMD), an inherited degenerative disease of the cardiac and skeletal muscle caused by absence of the protein dystrophin. We showed one hallmark of DMD cardiomyopathy is the dysregulation of cardiac gap junction channel protein connexin-43 (Cx43). Proper Cx43 localization and function at the cardiac intercalated disc (ID) is regulated by post-translational phosphorylation of Cx43-carboxy-terminus residues S325/S328/S330 (pS-Cx43). Concurrently, Cx43 traffics along microtubules (MTs) for targeted delivery to the ID. In DMD hearts, absence of dystrophin results in a hyperdensified and disorganized MT cytoskeleton, yet the link with pS-Cx43 remains unaddressed. To gain insight into the relationship between MTs and pS-Cx43, DMD mice (mdx) and pS-Cx43-deficient (mdxS3A) mice were treated with an inhibitor of MT polymerization, colchicine (Colch). Colch treatment protected mdx, not mdxS3A mice, against Cx43 remodeling, improved MT directionality, and enhanced pS-Cx43/tubulin interaction. Likewise, severe arrhythmias were prevented in isoproterenol-stressed mdx, not mdxS3A mice. Furthermore, MT directionality was improved in pS-Cx43-mimicking mdx (mdxS3E). Mdxutr+/- and mdxutr+/-S3A mice, lacking one copy of dystrophin homolog utrophin, displayed enhanced cardiac fibrosis and reduced lifespan compared with mdxutr+/-S3E; and Colch treatment corrected cardiac fibrosis in mdxutr+/- but not mdxutr+/-S3A. Collectively, the data suggest that improved MT directionality reduces Cx43 remodeling and that pS-Cx43 is necessary and sufficient to regulate MT organization, which plays crucial role in correcting cardiac dysfunction in DMD mice. Thus, identification of novel organizational mechanisms acting on pS-Cx43-MT will help develop novel cardioprotective therapies for DMD cardiomyopathy.NEW & NOTEWORTHY We found that colchicine administration to Cx43-phospho-deficient dystrophic mice fails to protect against Cx43 remodeling. Conversely, Cx43-phospho-mimic dystrophic mice display a normalized MT network. We envision a bidirectional regulation whereby correction of the dystrophic MTs leads to correction of Cx43 remodeling, which in turn leads to further correction of the MTs. Our findings suggest a link between phospho-Cx43 and MTs that provides strong foundations for novel therapeutics in DMD cardiomyopathy.
Collapse
Affiliation(s)
- Eric Himelman
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Julie Nouet
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Mauricio A Lillo
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Alexander Chong
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Delong Zhou
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Xander H T Wehrens
- Department of Molecular Physiology and Biophysics, Medicine, Neuroscience, and Pediatrics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Medicine, Neuroscience, and Pediatrics, Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas
| | - Lai-Hua Xie
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Natalia Shirokova
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Jorge E Contreras
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey
| |
Collapse
|
33
|
A Nonsense Variant in the DMD Gene Causes X-Linked Muscular Dystrophy in the Maine Coon Cat. Animals (Basel) 2022; 12:ani12212928. [PMID: 36359052 PMCID: PMC9653713 DOI: 10.3390/ani12212928] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Feline dystrophin-deficient muscular dystrophy (ddMD) is a fatal disease characterized by progressive weakness and degeneration of skeletal muscles and is caused by variants in the DMD gene. To date, only two feline causal variants have been identified. This study reports two cases of male Maine coon siblings that presented with muscular hypertrophy, growth retardation, weight loss, and vomiting. (2) Both cats were clinically examined and histopathology and immunofluorescent staining of the affected muscle was performed. DMD mRNA was sequenced to identify putative causal variants. (3) Both cats showed a significant increase in serum creatine kinase activity. Electromyography and histopathological examination of the muscle samples revealed abnormalities consistent with a dystrophic phenotype. Immunohistochemical testing revealed the absence of dystrophin, confirming the diagnosis of dystrophin-deficient muscular dystrophy. mRNA sequencing revealed a nonsense variant in exon 11 of the feline DMD gene, NC_058386.1 (XM_045050794.1): c.1180C > T (p.(Arg394*)), which results in the loss of the majority of the dystrophin protein. Perfect X-linked segregation of the variant was established in the pedigree. (4) ddMD was described for the first time in the Maine coon and the c.1180C>T variant was confirmed as the causal variant.
Collapse
|
34
|
Criscione J, Rezaei Z, Hernandez Cantu CM, Murphy S, Shin SR, Kim DH. Heart-on-a-chip platforms and biosensor integration for disease modeling and phenotypic drug screening. Biosens Bioelectron 2022; 220:114840. [DOI: 10.1016/j.bios.2022.114840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/09/2022] [Accepted: 10/18/2022] [Indexed: 11/02/2022]
|
35
|
Alheib O, da Silva LP, Kwon IK, Reis RL, Correlo VM. Preclinical research studies for treating severe muscular injuries: focus on tissue-engineered strategies. Trends Biotechnol 2022; 41:632-652. [PMID: 36266101 DOI: 10.1016/j.tibtech.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
Severe skeletal muscle injuries are a lifelong trauma with limited medical solutions. Significant progress has been made in developing in vitro surrogates for treating such trauma. However, more attention is needed when translating these approaches to the clinic. In this review, we survey the potential of tissue-engineered surrogates in promoting muscle healing, by critically analyzing data from recent preclinical models. The therapeutic advantages provided by a combination of different biomaterials, cell types, and biochemical mediators are discussed. Current therapies on muscle healing are also summarized, emphasizing their main advantages and drawbacks. We also discuss previous and ongoing clinical trials as well as highlighting future directions for the field.
Collapse
Affiliation(s)
- Omar Alheib
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lucília P da Silva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal; Department of Dental Materials, School of Dentistry, Kyung Hee University, Dongdaemun-gu, Seoul, Republic of Korea
| | - Vitor M Correlo
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
36
|
Den Hartog L, Asakura A. Implications of notch signaling in duchenne muscular dystrophy. Front Physiol 2022; 13:984373. [PMID: 36237531 PMCID: PMC9553129 DOI: 10.3389/fphys.2022.984373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
This review focuses upon the implications of the Notch signaling pathway in muscular dystrophies, particularly Duchenne muscular dystrophy (DMD): a pervasive and catastrophic condition concerned with skeletal muscle degeneration. Prior work has defined the pathogenesis of DMD, and several therapeutic approaches have been undertaken in order to regenerate skeletal muscle tissue and ameliorate the phenotype. There is presently no cure for DMD, but a promising avenue for novel therapies is inducing muscle regeneration via satellite cells (muscle stem cells). One specific target using this approach is the Notch signaling pathway. The canonical Notch signaling pathway has been well-characterized and it ultimately governs cell fate decision, cell proliferation, and induction of differentiation. Additionally, inhibition of the Notch signaling pathway has been directly implicated in the deficits seen with muscular dystrophies. Here, we explore the connection between the Notch signaling pathway and DMD, as well as how Notch signaling may be targeted to improve the muscle degeneration seen in muscular dystrophies.
Collapse
|
37
|
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: 12] [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
|
38
|
Ritter P, Nübler S, Buttgereit A, Smith LR, Mühlberg A, Bauer J, Michael M, Kreiß L, Haug M, Barton E, Friedrich O. Myofibrillar Lattice Remodeling Is a Structural Cytoskeletal Predictor of Diaphragm Muscle Weakness in a Fibrotic mdx ( mdx Cmah-/-) Model. Int J Mol Sci 2022; 23:ijms231810841. [PMID: 36142754 PMCID: PMC9500669 DOI: 10.3390/ijms231810841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a degenerative genetic myopathy characterized by complete absence of dystrophin. Although the mdx mouse lacks dystrophin, its phenotype is milder compared to DMD patients. The incorporation of a null mutation in the Cmah gene led to a more DMD-like phenotype (i.e., more fibrosis). Although fibrosis is thought to be the major determinant of ‘structural weakness’, intracellular remodeling of myofibrillar geometry was shown to be a major cellular determinant thereof. To dissect the respective contribution to muscle weakness, we assessed biomechanics and extra- and intracellular architecture of whole muscle and single fibers from extensor digitorum longus (EDL) and diaphragm. Despite increased collagen contents in both muscles, passive stiffness in mdx Cmah−/− diaphragm was similar to wt mice (EDL muscles were twice as stiff). Isometric twitch and tetanic stresses were 50% reduced in mdx Cmah−/− diaphragm (15% in EDL). Myofibrillar architecture was severely compromised in mdx Cmah−/− single fibers of both muscle types, but more pronounced in diaphragm. Our results show that the mdx Cmah−/− genotype reproduces DMD-like fibrosis but is not associated with changes in passive visco-elastic muscle stiffness. Furthermore, detriments in active isometric force are compatible with the pronounced myofibrillar disarray of the dystrophic background.
Collapse
Affiliation(s)
- Paul Ritter
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- Correspondence:
| | - Stefanie Nübler
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Andreas Buttgereit
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas R. Smith
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, CA 95618, USA
| | - Alexander Mühlberg
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Julian Bauer
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Mena Michael
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Lucas Kreiß
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Michael Haug
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
| | - Elisabeth Barton
- College of Health & Human Performance, University of Florida, Gainesville, FL 32611, USA
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Department of Chemical and Biological Engineering, Friedrich-Alexander-University Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052 Erlangen, Germany
- School of Medical Sciences, University of New South Wales, Wallace Wurth Building, 18 High Str., Sydney, NSW 2052, Australia
| |
Collapse
|
39
|
A Novel Method for Detecting Duchenne Muscular Dystrophy in Blood Serum of mdx Mice. Genes (Basel) 2022; 13:genes13081342. [PMID: 36011258 PMCID: PMC9407179 DOI: 10.3390/genes13081342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy, typically affecting males in infancy. The disease causes progressive weakness and atrophy of skeletal muscles, with approximately 20,000 new cases diagnosed yearly. Currently, methods for diagnosing DMD are invasive, laborious, and unable to make accurate early detections. While there is no cure for DMD, there are limited treatments available for managing symptoms. As such, there is a crucial unmet need to develop a simple and non-invasive method for accurately detecting DMD as early as possible. Raman spectroscopy with chemometric analysis is shown to have the potential to fill this diagnostic need.
Collapse
|
40
|
Babaeijandaghi F, Cheng R, Kajabadi N, Soliman H, Chang CK, Smandych J, Tung LW, Long R, Ghassemi A, Rossi FMV. Metabolic reprogramming of skeletal muscle by resident macrophages points to CSF1R inhibitors as muscular dystrophy therapeutics. Sci Transl Med 2022; 14:eabg7504. [PMID: 35767650 DOI: 10.1126/scitranslmed.abg7504] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The role of tissue-resident macrophages during tissue regeneration or fibrosis is not well understood, mainly due to the lack of a specific marker for their identification. Here, we identified three populations of skeletal muscle-resident myelomonocytic cells: a population of macrophages positive for lymphatic vessel endothelial receptor 1 (LYVE1) and T cell membrane protein 4 (TIM4 or TIMD4), a population of LYVE1-TIM4- macrophages, and a population of cells likely representing dendritic cells that were positive for CD11C and major histocompatibility complex class II (MHCII). Using a combination of parabiosis and lineage-tracing experiments, we found that, at steady state, TIM4- macrophages were replenished from the blood, whereas TIM4+ macrophages locally self-renewed [self-renewing resident macrophages (SRRMs)]. We further showed that Timd4 could be reliably used to distinguish SRRMs from damage-induced infiltrating macrophages. Using a colony-stimulating factor 1 receptor (CSF1R) inhibition/withdrawal approach to specifically deplete SRRMs, we found that SRRMs provided a nonredundant function in clearing damage-induced apoptotic cells early after extensive acute injury. In contrast, in chronic mild injury as seen in a mouse model of Duchenne muscular dystrophy, depletion of both TIM4-- and TIM4+-resident macrophage populations through long-term CSF1R inhibition changed muscle fiber composition from damage-sensitive glycolytic fibers toward damage-resistant glycolytic-oxidative fibers, thereby protecting muscle against contraction-induced injury both ex vivo and in vivo. This work reveals a previously unidentified role for resident macrophages in modulating tissue metabolism and may have therapeutic potential given the ongoing clinical testing of CSF1R inhibitors.
Collapse
Affiliation(s)
- Farshad Babaeijandaghi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ryan Cheng
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Nasim Kajabadi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Hesham Soliman
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minia 61519, Egypt.,Aspect Biosystems, 1781 W 75th Ave, Vancouver, BC V6P 6P2, Canada
| | - Chih-Kai Chang
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Josh Smandych
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Lin Wei Tung
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Reece Long
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Amirhossein Ghassemi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Fabio M V Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
41
|
Wang R, Kumar B, Doud EH, Mosley AL, Alexander MS, Kunkel LM, Nakshatri H. Skeletal muscle-specific overexpression of miR-486 limits mammary tumor-induced skeletal muscle functional limitations. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 28:231-248. [PMID: 35402076 PMCID: PMC8971682 DOI: 10.1016/j.omtn.2022.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/12/2022] [Indexed: 11/28/2022]
Abstract
miR-486 is a myogenic microRNA, and its reduced skeletal muscle expression is observed in muscular dystrophy. Transgenic overexpression of miR-486 using muscle creatine kinase promoter (MCK-miR-486) partially rescues muscular dystrophy phenotype. We had previously demonstrated reduced circulating and skeletal muscle miR-486 levels with accompanying skeletal muscle defects in mammary tumor models. To determine whether skeletal muscle miR-486 is functionally similar in dystrophies and cancer, we performed functional limitations and biochemical studies of skeletal muscles of MMTV-Neu mice that mimic HER2+ breast cancer and MMTV-PyMT mice that mimic luminal subtype B breast cancer and these mice crossed to MCK-miR-486 mice. miR-486 significantly prevented tumor-induced reduction in muscle contraction force, grip strength, and rotarod performance in MMTV-Neu mice. In this model, miR-486 reversed cancer-induced skeletal muscle changes, including loss of p53, phospho-AKT, and phospho-laminin alpha 2 (LAMA2) and gain of hnRNPA0 and SRSF10 phosphorylation. LAMA2 is a part of the dystrophin-associated glycoprotein complex, and its loss of function causes congenital muscular dystrophy. Complementing these beneficial effects on muscle, miR-486 indirectly reduced tumor growth and improved survival, which is likely due to systemic effects of miR-486 on production of pro-inflammatory cytokines such as IL-6. Thus, similar to dystrophy, miR-486 has the potential to reverse skeletal muscle defects and cancer burden.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brijesh Kumar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Emma H. Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Amber L. Mosley
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew S. Alexander
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children’s of Alabama, Birmingham, AL 35294, USA
| | - Louis M. Kunkel
- Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Richard L Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
42
|
Karri DR, Zhang Y, Chemello F, Min YL, Huang J, Kim J, Mammen PP, Xu L, Liu N, Bassel-Duby R, Olson EN. Long-term maintenance of dystrophin expression and resistance to injury of skeletal muscle in gene edited DMD mice. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:154-167. [PMID: 35402069 PMCID: PMC8956962 DOI: 10.1016/j.omtn.2022.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/03/2022] [Indexed: 11/24/2022]
|
43
|
Song MH, Yoo J, Oh JG, Kook H, Park WJ, Jeong D. Matricellular Protein CCN5 Gene Transfer Ameliorates Cardiac and Skeletal Dysfunction in mdx/utrn (±) Haploinsufficient Mice by Reducing Fibrosis and Upregulating Utrophin Expression. Front Cardiovasc Med 2022; 9:763544. [PMID: 35557546 PMCID: PMC9088811 DOI: 10.3389/fcvm.2022.763544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/31/2022] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration due to dystrophin gene mutations. Patients with DMD initially experience muscle weakness in their limbs during adolescence. With age, patients develop fatal respiratory and cardiac dysfunctions. During the later stages of the disease, severe cardiac fibrosis occurs, compromising cardiac function. Previously, our research showed that the matricellular protein CCN5 has antifibrotic properties. Therefore, we hypothesized that CCN5 gene transfer would ameliorate cardiac fibrosis and thus improve cardiac function in DMD-induced cardiomyopathy. We utilized mdx/utrn (±) haploinsufficient mice that recapitulated the DMD-disease phenotypes and used an adeno-associated virus serotype-9 viral vector for CCN5 gene transfer. We evaluated the onset of cardiac dysfunction using echocardiography and determined the experimental starting point in 13-month-old mice. Two months after CCN5 gene transfer, cardiac function was significantly enhanced, and cardiac fibrosis was ameliorated. Additionally, running performance was improved in CCN5 gene-transfected mice. Furthermore, in silico gene profiling analysis identified utrophin as a novel transcriptional target of CCN5. This was supplemented by a utrophin promoter assay and RNA-seq analysis, which confirmed that CCN5 was directly associated with utrophin expression. Our results showed that CCN5 may be a promising therapeutic molecule for DMD-induced cardiac and skeletal dysfunction.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Gwangju, South Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Dongtak Jeong
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| |
Collapse
|
44
|
Effects of Low-Intensity and Long-Term Aerobic Exercise on the Psoas Muscle of mdx Mice: An Experimental Model of Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23094483. [PMID: 35562874 PMCID: PMC9105402 DOI: 10.3390/ijms23094483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscle disease characterized by the absence of the protein dystrophin, which causes a loss of sarcolemma integrity, determining recurrent muscle injuries, decrease in muscle function, and progressive degeneration. Currently, there is a need for therapeutic treatments to improve the quality of life of DMD patients. Here, we investigated the effects of a low-intensity aerobic training (37 sessions) on satellite cells, peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α protein (PGC-1α), and different types of fibers of the psoas muscle from mdx mice (DMD experimental model). Wildtype and mdx mice were randomly divided into sedentary and trained groups (n = 24). Trained animals were subjected to 37 sessions of low-intensity running on a motorized treadmill. Subsequently, the psoas muscle was excised and analyzed by immunofluorescence for dystrophin, satellite cells, myosin heavy chain (MHC), and PGC-1α content. The minimal Feret’s diameters of the fibers were measured, and light microscopy was applied to observe general morphological features of the muscles. The training (37 sessions) improved morphological features in muscles from mdx mice and caused an increase in the number of quiescent/activated satellite cells. It also increased the content of PGC-1α in the mdx group. We concluded that low-intensity aerobic exercise (37 sessions) was able to reverse deleterious changes determined by DMD.
Collapse
|
45
|
Stirm M, Fonteyne LM, Shashikadze B, Stöckl JB, Kurome M, Keßler B, Zakhartchenko V, Kemter E, Blum H, Arnold GJ, Matiasek K, Wanke R, Wurst W, Nagashima H, Knieling F, Walter MC, Kupatt C, Fröhlich T, Klymiuk N, Blutke A, Wolf E. Pig models for Duchenne muscular dystrophy – from disease mechanisms to validation of new diagnostic and therapeutic concepts. Neuromuscul Disord 2022; 32:543-556. [DOI: 10.1016/j.nmd.2022.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/22/2022] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
|
46
|
Description of Osmolyte Pathways in Maturing Mdx Mice Reveals Altered Levels of Taurine and Sodium/Myo-Inositol Co-Transporters. Int J Mol Sci 2022; 23:ijms23063251. [PMID: 35328671 PMCID: PMC8955384 DOI: 10.3390/ijms23063251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/11/2022] [Accepted: 03/13/2022] [Indexed: 01/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration. Osmotic stress participates to DMD pathology and altered levels of osmolyte pathway members have been reported. The goal of this study was to gain insight in osmoregulatory changes in the mdx mouse model by examining the expression of osmolyte pathway members, including taurine transporter (TauT), sodium myo-inositol co-transporter (SMIT), betaine GABA transporter (BGT), and aldose reductase (AR) in the skeletal muscles and diaphragm of mdx mice aged 4, 8, 12, and 26 weeks. Necrosis was most prominent in 12 week-old mdx mice, whereas the amount of regenerated fibers increased until week 26 in the tibialis anterior. TauT protein levels were downregulated in the tibialis anterior and gastrocnemius of 4 to 12 week-old mdx mice, but not in 26 week-old mice, whereas TauT levels in the diaphragm remained significantly lower in 26 week-old mdx mice. In contrast, SMIT protein levels were significantly higher in the muscles of mdx mice when compared to controls. Our study revealed differential regulation of osmolyte pathway members in mdx muscle, which points to their complex involvement in DMD pathogenesis going beyond general osmotic stress responses. These results highlight the potential of osmolyte pathway members as a research interest and future therapeutic target in dystrophinopathy.
Collapse
|
47
|
Shirakawa T, Ikushima A, Maruyama N, Nambu Y, Awano H, Osawa K, Nirasawa K, Negishi Y, Nishio H, Fukushima S, Matsuo M. A sandwich ELISA kit reveals marked elevation of titin N-terminal fragment levels in the urine of mdx mice. Animal Model Exp Med 2022; 5:48-55. [PMID: 35229992 PMCID: PMC8879618 DOI: 10.1002/ame2.12204] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The mdx mouse is a model of Duchenne muscular dystrophy (DMD), a fatal progressive muscle wasting disease caused by dystrophin deficiency, and is used most widely in preclinical studies. Mice with dystrophin deficiency, however, show milder muscle strength phenotypes than humans. In human, the introduction of a sandwich enzyme-linked immunosorbent assay (ELISA) kit revealed a more than 700-fold increase in titin N-terminal fragment levels in the urine of pediatric patients with DMD. Notably, the urinary titin level declines with aging, reflecting progression of muscle wasting. In mouse, development of a highly sensitive ELISA kit has been awaited. Here, a sandwich ELISA kit to measure titin N-terminal fragment levels in mouse urine was developed. The developed kit showed good linearity, recovery, and repeatability in measuring recombinant or natural mouse titin N-terminal fragment levels. The titin N-terminal fragment concentration in the urine of mdx mice was more than 500-fold higher than that of normal mice. Urinary titin was further analyzed by extending the collection of urine samples to both young (3-11 weeks old) and aged (56-58 weeks old) mdx mice. The concentration in the young group was significantly higher than that in the aged group. It was concluded that muscle protein breakdown is active and persistent in mdx mice even though the muscle phenotype is mild. Our results provide an opportunity to develop DMD treatments that aim to alleviate muscle protein breakdown by monitoring urinary titin levels.
Collapse
Affiliation(s)
- Taku Shirakawa
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
- KNC Department of Nucleic Acid Drug DiscoveryFaculty of RehabilitationKobe Gakuin UniversityKobeJapan
| | - Ayumu Ikushima
- Department of PharmaceuticsFaculty of Pharmaceutical SciencesKobe Gakuin UniversityKobeJapan
| | - Nobuhiro Maruyama
- Diagnostic & Research Reagents DivisionImmuno‐Biological Laboratories Co., LtdFujiokaJapan
| | - Yoshinori Nambu
- Department of PediatricsKobe University Graduate School of MedicineKobeJapan
| | - Hiroyuki Awano
- Department of PediatricsKobe University Graduate School of MedicineKobeJapan
| | - Kayo Osawa
- Department of Medical TechnologyFaculty of Health SciencesKobe Tokiwa UniversityKobeJapan
| | - Kei Nirasawa
- Department of Drug Delivery and Molecular BiopharmaceuticsSchool of PharmacyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular BiopharmaceuticsSchool of PharmacyTokyo University of Pharmacy and Life SciencesTokyoJapan
| | - Hisahide Nishio
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
| | - Shoji Fukushima
- Department of PharmaceuticsFaculty of Pharmaceutical SciencesKobe Gakuin UniversityKobeJapan
| | - Masafumi Matsuo
- Research Center for Locomotion BiologyKobe Gakuin UniversityKobeJapan
- KNC Department of Nucleic Acid Drug DiscoveryFaculty of RehabilitationKobe Gakuin UniversityKobeJapan
| |
Collapse
|
48
|
Gartz M, Haberman M, Prom MJ, Beatka MJ, Strande JL, Lawlor MW. A Long-Term Study Evaluating the Effects of Nicorandil Treatment on Duchenne Muscular Dystrophy-Associated Cardiomyopathy in mdx Mice. J Cardiovasc Pharmacol Ther 2022; 27:10742484221088655. [PMID: 35353647 DOI: 10.1177/10742484221088655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by dystrophin gene mutations affecting striated muscle. Due to advances in skeletal muscle treatment, cardiomyopathy has emerged as a leading cause of death. Previously, nicorandil, a drug with antioxidant and nitrate-like properties, ameliorated cardiac damage and improved cardiac function in young, injured mdx mice. Nicorandil mitigated damage by stimulating antioxidant activity and limiting pro-oxidant expression. Here, we examined whether nicorandil was similarly cardioprotective in aged mdx mice. METHODS AND RESULTS Nicorandil (6 mg/kg) was given over 15 months. Echocardiography of mdx mice showed some functional defects at 12 months compared to wild-type (WT) mice, but not at 15 months. Disease manifestation was evident in mdx mice via treadmill assays and survival, but not open field and grip strength assays. Cardiac levels of SOD2 and NOX4 were decreased in mdx vs. WT. Nicorandil increased survival in mdx but did not alter cardiac function, fibrosis, diaphragm function or muscle fatigue. CONCLUSIONS In contrast to our prior work in young, injured mdx mice, nicorandil did not exert cardioprotective effects in 15 month aged mdx mice. Discordant findings may be explained by the lack of cardiac disease manifestation in aged mdx mice compared to WT, whereas significant cardiac dysfunction was previously seen with the sub-acute injury in young mice. Therefore, we are not able to conclude any cardioprotective effects with long-term nicorandil treatment in aging mdx mice.
Collapse
Affiliation(s)
- Melanie Gartz
- Department of Cell Biology, Neurobiology and Anatomy, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Cardiovascular Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Neuroscience Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pathology and Laboratory Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| | - Margaret Haberman
- Cardiovascular Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Neuroscience Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pathology and Laboratory Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| | - Mariah J Prom
- Neuroscience Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pathology and Laboratory Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| | - Margaret J Beatka
- Neuroscience Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pathology and Laboratory Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jennifer L Strande
- Cardiovascular Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael W Lawlor
- Neuroscience Research Center, 5506Medical College of Wisconsin, Milwaukee, WI, USA.,Department of Pathology and Laboratory Medicine, 5506Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
49
|
Stirm M, Fonteyne LM, Shashikadze B, Lindner M, Chirivi M, Lange A, Kaufhold C, Mayer C, Medugorac I, Kessler B, Kurome M, Zakhartchenko V, Hinrichs A, Kemter E, Krause S, Wanke R, Arnold GJ, Wess G, Nagashima H, de Angelis MH, Flenkenthaler F, Kobelke LA, Bearzi C, Rizzi R, Bähr A, Reese S, Matiasek K, Walter MC, Kupatt C, Ziegler S, Bartenstein P, Fröhlich T, Klymiuk N, Blutke A, Wolf E. A scalable, clinically severe pig model for Duchenne muscular dystrophy. Dis Model Mech 2021; 14:273744. [PMID: 34796900 PMCID: PMC8688409 DOI: 10.1242/dmm.049285] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/11/2021] [Indexed: 11/20/2022] Open
Abstract
Large animal models for Duchenne muscular dystrophy (DMD) are crucial for evaluation of diagnostic procedures and treatment strategies. Pigs cloned from male cells lacking DMD exon 52 (DMDΔ52) resemble molecular, clinical and pathological hallmarks of DMD, but die before sexual maturity and cannot be propagated by breeding. Therefore, we generated female DMD+/- carriers. A single founder animal had 11 litters with 29 DMDY/-, 34 DMD+/- as well as 36 male and 29 female wild-type offspring. Breeding with F1 and F2 DMD+/- carriers resulted in additional 114 DMDY/- piglets. With intensive neonatal management, the majority survived for 3-4 months, providing statistically relevant cohorts for experimental studies. Pathological investigations and proteome studies of skeletal muscles and myocardium confirmed the resemblance of human disease mechanisms. Importantly, DMDY/- pigs reveal progressive myocardial fibrosis and increased expression of connexin-43, associated with significantly reduced left ventricular ejection fraction already at age 3 months. Furthermore, behavioral tests provided evidence for impaired cognitive ability. Our breeding cohort of DMDΔ52 pigs and standardized tissue repositories provide important resources for studying DMD disease mechanisms and for testing novel treatment strategies.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Lina Marie Fonteyne
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Magdalena Lindner
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Maila Chirivi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Andreas Lange
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Clara Kaufhold
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Christian Mayer
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Ivica Medugorac
- Population Genomics Group, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Barbara Kessler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Mayuko Kurome
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Valeri Zakhartchenko
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Arne Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Sabine Krause
- Friedrich Baur Institute, Department of Neurology, LMU Munich, Munich, Germany
| | - Rüdiger Wanke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Georg J Arnold
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Gerhard Wess
- Clinic of Small Animal Medicine, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki, Japan
| | | | - Florian Flenkenthaler
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Levin Arne Kobelke
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Institute of Genetic and Biomedical Research, UOS of Milan, National Research Council (IRGB-CNR), Milan, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, Milan, Italy.,Institute for Biomedical Technologies, National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Andrea Bähr
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Sven Reese
- Chair for Anatomy, Histology and Embryology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Kaspar Matiasek
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich Baur Institute, Department of Neurology, LMU Munich, Munich, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich and German Center for Cardiovascular Research (DZHK), Munich Heart Alliance, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| | - Nikolai Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Andreas Blutke
- Institute of Experimental Genetics, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich, Germany.,Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany.,Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich, Germany
| |
Collapse
|
50
|
Atmanli A, Chai AC, Cui M, Wang Z, Nishiyama T, Bassel-Duby R, Olson EN. Cardiac Myoediting Attenuates Cardiac Abnormalities in Human and Mouse Models of Duchenne Muscular Dystrophy. Circ Res 2021; 129:602-616. [PMID: 34372664 PMCID: PMC8416801 DOI: 10.1161/circresaha.121.319579] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Ayhan Atmanli
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andreas C. Chai
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Miao Cui
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Zhaoning Wang
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Takahiko Nishiyama
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eric N. Olson
- Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|