1
|
Zhang X, Zhao M, Deng P, Liu H, Li H, Zhang X, Wang D, Qin J. Modeling Idiopathic Inflammatory Myopathy in the Bioinspired Muscle Tissue on Chip. Adv Healthc Mater 2025:e2405111. [PMID: 40357815 DOI: 10.1002/adhm.202405111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/21/2025] [Indexed: 05/15/2025]
Abstract
Idiopathic inflammatory myopathy (IIM) is an autoimmune disease that characterized by non-purulent inflammation of the skeletal muscle. However, due to the limitation of study model that can recapitulate the complex pathological process of IIM, the pathogenesis of IIM is still not fully clear. This manuscript develops a vascularized muscle tissue model on a chip that allows to model the immunity mediated pathological changes in IIM. This vascularized muscle model is constructed by layer-by-layer assembly, which could coculture of endothelial cells, myoblasts, and monocytes in a perfusable 3D system. The vascularized muscle model exhibits good biofunctions, including muscle cells alignment and fusion, myofibers generation, force production and expression of muscular biomarkers (myosin heavy chains 1, myosin heavy chains 7, actinin alpha 2, myogenin, and Desmin). Exposure to perfusion of activated monocytes, this work observes the functional changes of muscle tissue, which referred to myofibers atrophy, inflammatory response, and downregulated expression of muscle mature marker, consistent with clinical features of IIM. This work provides a unique platform for modelling IIM and paves a promising avenue for myopathies study and drug testing.
Collapse
Affiliation(s)
- Xu Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Mengqian Zhao
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Pengwei Deng
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
| | - Haitao Liu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Hongjing Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Xiaoqing Zhang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Daqing Wang
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116000, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- University of Science and Technology of China, Hefei, 230026, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, 215123, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
2
|
Schreiber J, Rotard L, Tourneur Y, Lafoux A, Berthier C, Allard B, Huchet C, Jacquemond V. Reduced voltage-activated Ca2+ release flux in muscle fibers from a rat model of Duchenne dystrophy. J Gen Physiol 2025; 157:e202413588. [PMID: 39718509 DOI: 10.1085/jgp.202413588] [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: 04/09/2024] [Revised: 08/14/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
The potential pathogenic role of disturbed Ca2+ homeostasis in Duchenne muscular dystrophy (DMD) remains a complex, unsettled issue. We used muscle fibers isolated from 3-mo-old DMDmdx rats to further investigate the case. Most DMDmdx fibers exhibited no sign of trophic or morphology distinction as compared with WT fibers and mitochondria and t-tubule membrane networks also showed no stringent discrepancy. Under voltage clamp, values for holding current were similar in the two groups, whereas values for capacitance were larger in DMDmdx fibers, suggestive of enhanced amount of t-tubule membrane. The Ca2+ current density across the channel carried by the EC coupling voltage sensor (CaV1.1) was unchanged. The maximum rate of voltage-activated sarcoplasmic reticulum (SR) Ca2+ release was reduced by 25% in the DMDmdx fibers, with no change in voltage dependency. Imaging resting Ca2+ revealed rare spontaneous local SR Ca2+ release events with no sign of elevated activity in DMDmdx fibers. Under current clamp, DMDmdx fibers generated similar trains of action potentials as WT fibers. Results suggest that reduced peak amplitude of SR Ca2+ release is an inherent feature of this DMD model, likely contributing to muscle weakness. This occurs despite a preserved amount of releasable Ca2+ and with no change in excitability, CaV1.1 channel activity, and SR Ca2+ release at rest. Although we cannot exclude that fibers from the 3-mo-old animals do not yet display a fully developed disease phenotype, results provide limited support for pathomechanistic concepts frequently associated with DMD such as membrane fragility, excessive Ca2+ entry, or enhanced SR Ca2+ leak.
Collapse
Affiliation(s)
- Jonathan Schreiber
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Ludivine Rotard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Yves Tourneur
- UFPE Department Nutrição, Cidade Universitária, Recife, Brazil
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
| | - Christine Berthier
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Bruno Allard
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| | - Corinne Huchet
- Therassay Platform, CAPACITES, Nantes Université , Nantes, France
- Nantes Gene Therapy Laboratory, Nantes Université, INSERM UMR TARGET 1089, Nantes, France
| | - Vincent Jacquemond
- University Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5261, INSERM U-1315, Institut NeuroMyoGène - Pathophysiology and Genetics of Neuron and Muscle , Lyon, France
| |
Collapse
|
3
|
Wetzlich B, Nyakundi BB, Yang J. Therapeutic applications and challenges in myostatin inhibition for enhanced skeletal muscle mass and functions. Mol Cell Biochem 2025; 480:1535-1553. [PMID: 39340593 PMCID: PMC11842502 DOI: 10.1007/s11010-024-05120-y] [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: 05/28/2024] [Accepted: 09/07/2024] [Indexed: 09/30/2024]
Abstract
Myostatin, a potent negative regulator of skeletal muscle mass, has garnered significant attention as a therapeutic target for muscle dystrophies. Despite extensive research and promising preclinical results, clinical trials targeting myostatin inhibition in muscle dystrophies have failed to yield substantial improvements in muscle function or fitness in patients. This review details the mechanisms behind myostatin's function and the various inhibitors that have been tested preclinically and clinically. It also examines the challenges encountered in clinical translation, including issues with drug specificity, differences in serum myostatin concentrations between animal models and humans, and the necessity of neural input for functional improvements. Additionally, we explore promising avenues of research beyond muscle dystrophies, particularly in the treatment of metabolic syndromes and orthopedic disorders. Insights from these alternative applications suggest that myostatin inhibition may hold the potential for addressing a broader range of pathologies, providing new directions for therapeutic development.
Collapse
Affiliation(s)
- Brock Wetzlich
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Benard B Nyakundi
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Jinzeng Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
4
|
Gómez Armengol E, Merckx C, De Sutter H, De Bleecker JL, De Paepe B. Changes to the Autophagy-Related Muscle Proteome Following Short-Term Treatment with Ectoine in the Duchenne Muscular Dystrophy Mouse Model mdx. Int J Mol Sci 2025; 26:439. [PMID: 39859157 PMCID: PMC11765399 DOI: 10.3390/ijms26020439] [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: 11/15/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
The most severe form of muscular dystrophy (MD), known as Duchenne MD (DMD), remains an incurable disease, hence the ongoing efforts to develop supportive therapies. The dysregulation of autophagy, a degradative yet protective mechanism activated when tissues are under severe and prolonged stress, is critically involved in DMD. Treatments that harness autophagic capacities therefore represent a promising therapeutic approach. Osmolytes are protective organic molecules that regulate osmotic pressure and cellular homeostasis and may support tissue-repairing autophagy. We therefore explored the effects of the osmolyte ectoine in the standard mouse model of DMD, the mdx, focusing on the autophagy-related proteome. Mice were treated with ectoine in their drinking water (150 mg/kg) or through daily intraperitoneal injection (177 mg/kg) until they were 5.5 weeks old. Hind limb muscles were dissected, and samples were prepared for Western blotting for protein quantification and for immunofluorescence for an evaluation of tissue distribution. We report changes in the protein levels of autophagy-related 5 (ATG5), Ser366-phosphorylated sequestosome 1 (SQSTM1), heat shock protein 70 (HSP70), activated microtubule-associated protein 1A/1B-light chain 3 (LC3 II) and mammalian target of rapamycin (mTOR). Most importantly, ectoine significantly improved the balance between LC3 II and SQSTM1 levels in mdx gastrocnemius muscle, and LC3 II immunostaining was most pronounced in muscle fibers of the tibialis anterior from treated mdx. These findings lend support for the further investigation of ectoine as a potential therapeutic intervention for DMD.
Collapse
MESH Headings
- Animals
- Amino Acids, Diamino/pharmacology
- Amino Acids, Diamino/administration & dosage
- Autophagy/drug effects
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/pathology
- Mice, Inbred mdx
- Mice
- Disease Models, Animal
- Proteome/metabolism
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Male
- Sequestosome-1 Protein/metabolism
- HSP70 Heat-Shock Proteins/metabolism
- Mice, Inbred C57BL
Collapse
Affiliation(s)
| | | | | | | | - Boel De Paepe
- Neuromuscular Reference Center and Department of Neurology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium (J.L.D.B.)
| |
Collapse
|
5
|
Otake M, Imamura M, Enya S, Kangawa A, Shibata M, Ozaki K, Kimura K, Ono E, Aoki Y. Severe cardiac and skeletal manifestations in DMD-edited microminipigs: an advanced surrogate for Duchenne muscular dystrophy. Commun Biol 2024; 7:523. [PMID: 38702481 PMCID: PMC11068776 DOI: 10.1038/s42003-024-06222-5] [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: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 05/06/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is an intractable X-linked muscular dystrophy caused by mutations in the DMD gene. While many animal models have been used to study the disease, translating findings to humans has been challenging. Microminipigs, with their pronounced physiological similarity to humans and notably compact size amongst pig models, could offer a more representative model for human diseases. Here, we accomplished precise DMD modification in microminipigs by co-injecting embryos with Cas9 protein and a single-guide RNA targeting exon 23 of DMD. The DMD-edited microminipigs exhibited pronounced clinical phenotypes, including perturbed locomotion and body-wide skeletal muscle weakness and atrophy, alongside augmented serum creatine kinase levels. Muscle weakness was observed as of one month of age, respiratory and cardiac dysfunctions emerged by the sixth month, and the maximum lifespan was 29.9 months. Histopathological evaluations confirmed dystrophin deficiency and pronounced dystrophic pathology in the skeletal and myocardial tissues, demonstrating that these animals are an unprecedented model for studying human DMD. The model stands as a distinct and crucial tool in biomedical research, offering deep understanding of disease progression and enhancing therapeutic assessments, with potential to influence forthcoming treatment approaches.
Collapse
Affiliation(s)
- Masayoshi Otake
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, Kikugawa, Shizuoka, 439-0037, Japan.
| | - Michihiro Imamura
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, 187-8502, Japan
| | - Satoko Enya
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, Kikugawa, Shizuoka, 439-0037, Japan
| | - Akihisa Kangawa
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, Kikugawa, Shizuoka, 439-0037, Japan
| | - Masatoshi Shibata
- Swine and Poultry Research Center, Shizuoka Prefectural Research Institute of Animal Industry, Kikugawa, Shizuoka, 439-0037, Japan
| | - Kinuyo Ozaki
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koichi Kimura
- Departments of Laboratory Medicine/Cardiology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsuro Ono
- Department of Biomedicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
- Center of Biomedical Research, Research Center for Human Disease Modeling, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Centre of Neurology and Psychiatry, Tokyo, 187-8502, Japan.
| |
Collapse
|
6
|
Wilpert NM, Tonduti D, Vaia Y, Krude H, Sarret C, Schuelke M. Establishing Patient-Centered Outcomes for MCT8 Deficiency: Stakeholder Engagement and Systematic Literature Review. Neuropsychiatr Dis Treat 2023; 19:2195-2216. [PMID: 37881807 PMCID: PMC10595182 DOI: 10.2147/ndt.s379703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 10/07/2023] [Indexed: 10/27/2023] Open
Abstract
Introduction The SCL16A2 gene encodes the thyroid hormone (TH) transporter MCT8. Pathogenic variants result in a reduced TH uptake into the CNS despite high serum T3 concentrations. Patients suffer from severe neurodevelopmental delay and require multidisciplinary care. Since a first compassionate use study in 2008, the development of therapies has recently gained momentum. Treatment strategies range from symptom-based approaches, supplementation with TH or TH-analogs, to gene therapy. All these studies have mainly used surrogate endpoints and clinical outcomes. However, the EMA and FDA strongly encourage researchers to involve patients and their advocacy groups in the design of clinical trials. This should strengthen the patients' perspective and identify clinical endpoints that are clinically relevant to their daily life. Methods We involved patient families to define patient-relevant outcomes for MCT8 deficiency. In close collaboration with patient families, we designed a questionnaire asking for their five most preferred therapeutic goals, which, if achieved at least, make a difference in their lives. In addition, we performed a systematic review according to Cochrane recommendations of the published treatment trials. Results We obtained results from 15 families with completed questionnaires from 14 mothers and 8 fathers. Improvement in development, especially in gross motor skills, was most important to the parents. 59% wished for head control and 50% for sitting ability. Another 36% wished for weight gain, 32% for improvement of expressive language skills, and 18% for a reduction of dystonia/spasticity, less dysphagia, and reflux. Paraclinical aspects were least important (5-9%). In a treatment trial (n=46) and compassionate use cases (n=83), the results were mainly inconclusive, partly due to a lack of predefined patient-centered clinical endpoints. Discussion We recommend that future trials should define a relevant improvement in "development" and/or other patient-relevant outcomes compared to natural history as treatment goals.
Collapse
Affiliation(s)
- Nina-Maria Wilpert
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Pediatric Neurology, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Center for Chronically Sick Children, Berlin, Germany
| | - Davide Tonduti
- Unit of Pediatric Neurology, C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children’s Hospital, Università Degli Studi Di Milano, Milan, Italy
| | - Ylenia Vaia
- Unit of Pediatric Neurology, C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), V. Buzzi Children’s Hospital, Università Degli Studi Di Milano, Milan, Italy
| | - Heiko Krude
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Experimental Pediatric Endocrinology, Berlin, Germany
| | - Catherine Sarret
- Centre de Compétence des Leucodystrophies et Leucoencéphalopathies de Cause Rare, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Markus Schuelke
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Department of Pediatric Neurology, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health (BIH), Center for Chronically Sick Children, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), NeuroCure Clinical Research Center, Berlin, Germany
| |
Collapse
|
7
|
Lemos JP, Tenório LPG, Mouly V, Butler-Browne G, Mendes-da-Cruz DA, Savino W, Smeriglio P. T cell biology in neuromuscular disorders: a focus on Duchenne Muscular Dystrophy and Amyotrophic Lateral Sclerosis. Front Immunol 2023; 14:1202834. [PMID: 37920473 PMCID: PMC10619758 DOI: 10.3389/fimmu.2023.1202834] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023] Open
Abstract
Growing evidence demonstrates a continuous interaction between the immune system, the nerve and the muscle in neuromuscular disorders of different pathogenetic origins, such as Duchenne Muscular Dystrophy (DMD) and Amyotrophic Lateral Sclerosis (ALS), the focus of this review. Herein we highlight the complexity of the cellular and molecular interactions involving the immune system in neuromuscular disorders, as exemplified by DMD and ALS. We describe the distinct types of cell-mediated interactions, such as cytokine/chemokine production as well as cell-matrix and cell-cell interactions between T lymphocytes and other immune cells, which target cells of the muscular or nervous tissues. Most of these interactions occur independently of exogenous pathogens, through ligand-receptor binding and subsequent signal transduction cascades, at distinct levels of specificity. Although this issue reveals the complexity of the system, it can also be envisioned as a window of opportunity to design therapeutic strategies (including synthetic moieties, cell and gene therapy, as well as immunotherapy) by acting upon one or more targets. In this respect, we discuss ongoing clinical trials using VLA-4 inhibition in DMD, and in ALS, with a focus on regulatory T cells, both revealing promising results.
Collapse
Affiliation(s)
- Julia Pereira Lemos
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Liliane Patrícia Gonçalves Tenório
- Laboratory of Cell Biology, Institute of Biological and Health Sciences, Federal University of Alagoas, Maceio, Alagoas, Brazil
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Vincent Mouly
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Gillian Butler-Browne
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation (RENEURIN), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Piera Smeriglio
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| |
Collapse
|
8
|
Miranda Alarcón YS, Jazwinska D, Lymon T, Khalili A, Browe D, Newton B, Pellegrini M, Cohen RI, Shreiber DI, Freeman JW. The Use of Collagen Methacrylate in Actuating Polyethylene Glycol Diacrylate-Acrylic Acid Scaffolds for Muscle Regeneration. Ann Biomed Eng 2023; 51:1165-1180. [PMID: 36853478 DOI: 10.1007/s10439-023-03139-8] [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: 05/25/2022] [Accepted: 01/03/2023] [Indexed: 03/01/2023]
Abstract
After muscle loss or injury, skeletal muscle tissue has the ability to regenerate and return its function. However, large volume defects in skeletal muscle tissue pose a challenge to regenerate due to the absence of regenerative elements such as biophysical and biochemical cues, making the development of new treatments necessary. One potential solution is to utilize electroactive polymers that can change size or shape in response to an external electric field. Poly(ethylene glycol) diacrylate (PEGDA) is one such polymer, which holds great potential as a scaffold for muscle tissue regeneration due to its mechanical properties. In addition, the versatile chemistry of this polymer allows for the conjugation of new functional groups to enhance its electroactive properties and biocompatibility. Herein, we have developed an electroactive copolymer of PEGDA and acrylic acid (AA) in combination with collagen methacrylate (CMA) to promote cell adhesion and proliferation. The electroactive properties of the CMA + PEGDA:AA constructs were investigated through actuation studies. Furthermore, the biological properties of the hydrogel were investigated in a 14-day in vitro study to evaluate myosin light chain (MLC) expression and metabolic activity of C2C12 mouse myoblast cells. The addition of CMA improved some aspects of material bioactivity, such as MLC expression in C2C12 mouse myoblast cells. However, the incorporation of CMA in the PEGDA:AA hydrogels reduced the sample movement when placed under an electric field, possibly due to steric hindrance from the CMA. Further research is needed to optimize the use of CMA in combination with PEGDA:AA as a potential scaffold for skeletal muscle tissue engineering.
Collapse
Affiliation(s)
| | - Dorota Jazwinska
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Terrence Lymon
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Amin Khalili
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Daniel Browe
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Brandon Newton
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Michael Pellegrini
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA
| | - Joseph W Freeman
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|
9
|
Dystrophin ( DMD) Missense Variant in Cats with Becker-Type Muscular Dystrophy. Int J Mol Sci 2023; 24:ijms24043192. [PMID: 36834603 PMCID: PMC9964367 DOI: 10.3390/ijms24043192] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/20/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Muscular dystrophy due to dystrophin deficiency in humans is phenotypically divided into a severe Duchenne and milder Becker type. Dystrophin deficiency has also been described in a few animal species, and few DMD gene variants have been identified in animals. Here, we characterize the clinical, histopathological, and molecular genetic aspects of a family of Maine Coon crossbred cats with clinically mild and slowly progressive muscular dystrophy. Two young adult male littermate cats exhibited abnormal gait and muscular hypertrophy with macroglossia. Serum creatine kinase activities were highly increased. Histopathologically, dystrophic skeletal muscle exhibited marked structural changes including atrophic, hypertrophic, and necrotic muscle fibers. Immunohistochemistry showed irregularly reduced expression of dystrophin but the staining of other muscle proteins such as β- and γ-sarcoglycans as well as desmin was also diminished. Whole genome sequencing of one affected cat and genotyping of the littermate found both to be hemizygous mutant at a single DMD missense variant (c.4186C>T). No other protein-changing variants in candidate genes for muscular dystrophy were detected. In addition, one clinically healthy male littermate was hemizygous wildtype, while the queen and one female littermate were clinically healthy, but heterozygous. The predicted amino acid exchange (p.His1396Tyr) resides in a conserved central rod spectrin domain of dystrophin. Various protein modeling programs did not predict major disruption of the dystrophin protein by this substitution, but the altered charge of the region may still affect protein function. This study represents the first genotype-to-phenotype correlation of Becker-type dystrophin deficiency in companion animals.
Collapse
|
10
|
Baumann CW, Ingalls CP, Lowe DA. Mechanisms of weakness in Mdx muscle following in vivo eccentric contractions. J Muscle Res Cell Motil 2022; 43:63-72. [PMID: 35445349 DOI: 10.1007/s10974-022-09617-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/30/2022] [Indexed: 02/03/2023]
Abstract
Skeletal muscle of the dystrophin-deficient mdx mouse is hypersensitive to eccentric (ECC) contraction-induced strength loss due to plasmalemmal electrical dysfunction. Despite plasmalemmal inexcitability being a logical mechanism responsible for weakness, it remains unclear if processes up- and/or down-stream remain functionally intact in injured mdx muscle. The purpose of this study was to analyze additional processes necessary for excitation-contraction coupling that are potentially disrupted by ECC contractions. Anterior crural muscles (tibialis anterior, extensor digitorum longus [EDL], and extensor hallucis muscles) of wildtype (WT) and mdx mice were injured in vivo with 50 ECC contractions and torque was measured immediately before and after the contraction bout. Following the in vivo assessment, EDL ex vivo isometric and caffeine forces were analyzed. In vivo isometric torque and ex vivo force in WT muscle were reduced 38 and 30% (p < 0.001), while caffeine force was also reduced (p = 0.021), albeit to a lesser degree (9%). In contrast, in vivo isometric torque, ex vivo isometric force and ex vivo caffeine-induced force were all reduced 56-67% (p < 0.001) in mdx muscle and did not differ from one another (p = 0.114). Disproportional reductions in isometric strength and caffeine-induced force confirm that ECC contractions uncoupled the plasmalemma from the ryanodine receptors (RyRs) in WT muscle. In mdx muscle, the proportional reductions in isometric strength and caffeine-induced force following ECC contractions reveal that dysfunction occurs at and/or distal to the RyRs immediately post-injury. Thus, weakness in injured mdx muscle cannot be isolated to one mechanism, rather several steps of muscle contraction are disrupted.
Collapse
Affiliation(s)
- Cory W Baumann
- Ohio Musculoskeletal and Neurological Institute (OMNI), Department of Biomedical Sciences, Ohio University, Athens, OH, USA.
| | - Christopher P Ingalls
- Department of Kinesiology and Health, Georgia State University, Atlanta, Georgia, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science, Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Micheli L, Mitidieri E, Turnaturi C, Vanacore D, Ciampi C, Lucarini E, Cirino G, Ghelardini C, Sorrentino R, Di Cesare Mannelli L, d’Emmanuele di Villa Bianca R. Beneficial Effect of H 2S-Releasing Molecules in an In Vitro Model of Sarcopenia: Relevance of Glucoraphanin. Int J Mol Sci 2022; 23:5955. [PMID: 35682634 PMCID: PMC9180606 DOI: 10.3390/ijms23115955] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Sarcopenia is a gradual and generalized skeletal muscle (SKM) syndrome, characterized by the impairment of muscle components and functionality. Hydrogen sulfide (H2S), endogenously formed within the body from the activity of cystathionine-γ-lyase (CSE), cystathionine- β-synthase (CBS), and mercaptopyruvate sulfurtransferase, is involved in SKM function. Here, in an in vitro model of sarcopenia based on damage induced by dexamethasone (DEX, 1 μM, 48 h treatment) in C2C12-derived myotubes, we investigated the protective potential of exogenous and endogenous sources of H2S, i.e., glucoraphanin (30 μM), L-cysteine (150 μM), and 3-mercaptopyruvate (150 μM). DEX impaired the H2S signalling in terms of a reduction in CBS and CSE expression and H2S biosynthesis. Glucoraphanin and 3-mercaptopyruvate but not L-cysteine prevented the apoptotic process induced by DEX. In parallel, the H2S-releasing molecules reduced the oxidative unbalance evoked by DEX, reducing catalase activity, O2- levels, and protein carbonylation. Glucoraphanin, 3-mercaptopyruvate, and L-cysteine avoided the changes in myotubes morphology and morphometrics after DEX treatment. In conclusion, in an in vitro model of sarcopenia, an impairment in CBS/CSE/H2S signalling occurs, whereas glucoraphanin, a natural H2S-releasing molecule, appears more effective for preventing the SKM damage. Therefore, glucoraphanin supplementation could be an innovative therapeutic approach in the management of sarcopenia.
Collapse
Affiliation(s)
- Laura Micheli
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba—Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.M.); (C.C.); (E.L.); (C.G.)
| | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.M.); (C.T.); (D.V.); (G.C.); (R.d.d.V.B.)
| | - Carlotta Turnaturi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.M.); (C.T.); (D.V.); (G.C.); (R.d.d.V.B.)
| | - Domenico Vanacore
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.M.); (C.T.); (D.V.); (G.C.); (R.d.d.V.B.)
| | - Clara Ciampi
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba—Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.M.); (C.C.); (E.L.); (C.G.)
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba—Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.M.); (C.C.); (E.L.); (C.G.)
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.M.); (C.T.); (D.V.); (G.C.); (R.d.d.V.B.)
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba—Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.M.); (C.C.); (E.L.); (C.G.)
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health-Neurofarba—Section of Pharmacology and Toxicology, University of Florence, 50139 Florence, Italy; (L.M.); (C.C.); (E.L.); (C.G.)
| | - Roberta d’Emmanuele di Villa Bianca
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy; (E.M.); (C.T.); (D.V.); (G.C.); (R.d.d.V.B.)
| |
Collapse
|
12
|
Stefano MED, Ferretti V, Mozzetta C. Synaptic alterations as a neurodevelopmental trait of Duchenne muscular dystrophy. Neurobiol Dis 2022; 168:105718. [PMID: 35390481 DOI: 10.1016/j.nbd.2022.105718] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 01/14/2023] Open
Abstract
Dystrophinopaties, e.g., Duchenne muscular dystrophy (DMD), Becker muscular dystrophy and X-linked dilated cardiomyopathy are inherited neuromuscular diseases, characterized by progressive muscular degeneration, which however associate with a significant impact on general system physiology. The more severe is the pathology and its diversified manifestations, the heavier are its effects on organs, systems, and tissues other than muscles (skeletal, cardiac and smooth muscles). All dystrophinopaties are characterized by mutations in a single gene located on the X chromosome encoding dystrophin (Dp427) and its shorter isoforms, but DMD is the most devasting: muscular degenerations manifests within the first 4 years of life, progressively affecting motility and other muscular functions, and leads to a fatal outcome between the 20s and 40s. To date, after years of studies on both DMD patients and animal models of the disease, it has been clearly demonstrated that a significant percentage of DMD patients are also afflicted by cognitive, neurological, and autonomic disorders, of varying degree of severity. The anatomical correlates underlying neural functional damages are established during embryonic development and the early stages of postnatal life, when brain circuits, sensory and motor connections are still maturing. The impact of the absence of Dp427 on the development, differentiation, and consolidation of specific cerebral circuits (hippocampus, cerebellum, prefrontal cortex, amygdala) is significant, and amplified by the frequent lack of one or more of its lower molecular mass isoforms. The most relevant aspect, which characterizes DMD-associated neurological disorders, is based on morpho-functional alterations of selective synaptic connections within the affected brain areas. This pathological feature correlates neurological conditions of DMD to other severe neurological disorders, such as schizophrenia, epilepsy and autistic spectrum disorders, among others. This review discusses the organization and the role of the dystrophin-dystroglycan complex in muscles and neurons, focusing on the neurological aspect of DMD and on the most relevant morphological and functional synaptic alterations, in both central and autonomic nervous systems, described in the pathology and its animal models.
Collapse
Affiliation(s)
- Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy.
| | - Valentina Ferretti
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy; Center for Research in Neurobiology Daniel Bovet, Sapienza University of Rome, 00185 Rome, Italy
| | - Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy c/o Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
13
|
MUNNO PAULAM, BARROSO POLIANAR, VASCONCELOS BRUNAF, SILVA GEOVANNEBDA, SALGUEIRO THIAGOM, AGUIAR HELOISAH, VITORINO LETÍCIAM, OLIVEIRA MURILOX, MARTINS HELENR, GAIAD THAISP, MACHADO ALEXS. Acute toxicity and regenerative dose finding of an extract of Miconia ferruginata DC. in a mouse model of Duchenne muscular dystrophy. AN ACAD BRAS CIENC 2022; 94:e20210190. [DOI: 10.1590/0001-3765202220210190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/02/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- PAULA M. MUNNO
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | | | | | | | | | | | | | | | - HELEN R. MARTINS
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | - THAIS P. GAIAD
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | | |
Collapse
|
14
|
Ebrahimi M, Lad H, Fusto A, Tiper Y, Datye A, Nguyen CT, Jacques E, Moyle LA, Nguyen T, Musgrave B, Chávez-Madero C, Bigot A, Chen C, Turner S, Stewart BA, Pegoraro E, Vitiello L, Gilbert PM. De novo revertant fiber formation and therapy testing in a 3D culture model of Duchenne muscular dystrophy skeletal muscle. Acta Biomater 2021; 132:227-244. [PMID: 34048976 DOI: 10.1016/j.actbio.2021.05.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022]
Abstract
The biological basis of Duchenne muscular dystrophy (DMD) pathology is only partially characterized and there are still few disease-modifying therapies available, therein underlying the value of strategies to model and study DMD. Dystrophin, the causative gene of DMD, is responsible for linking the cytoskeleton of muscle fibers to the extracellular matrix beyond the sarcolemma. We posited that disease-associated phenotypes not yet captured by two-dimensional culture methods would arise by generating multinucleated muscle cells within a three-dimensional (3D) extracellular matrix environment. Herein we report methods to produce 3D human skeletal muscle microtissues (hMMTs) using clonal, immortalized myoblast lines established from healthy and DMD donors. We also established protocols to evaluate immortalized hMMT self-organization and myotube maturation, as well as calcium handling, force generation, membrane stability (i.e., creatine kinase activity and Evans blue dye permeability) and contractile apparatus organization following electrical-stimulation. In examining hMMTs generated with a cell line wherein the dystrophin gene possessed a duplication of exon 2, we observed rare dystrophin-positive myotubes, which were not seen in 2D cultures. Further, we show that treating DMD hMMTs with a β1-integrin activating antibody, improves contractile apparatus maturation and stability. Hence, immortalized myoblast-derived DMD hMMTs offer a pre-clinical system with which to investigate the potential of duplicated exon skipping strategies and those that protect muscle cells from contraction-induced injury. STATEMENT OF SIGNIFICANCE: Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disorder that is caused by mutation of the dystrophin gene. The biological basis of DMD pathology is only partially characterized and there is no cure for this fatal disease. Here we report a method to produce 3D human skeletal muscle microtissues (hMMTs) using immortalized human DMD and healthy myoblasts. Morphological and functional assessment revealed DMD-associated pathophysiology including impaired calcium handling and de novo formation of dystrophin-positive revertant muscle cells in immortalized DMD hMMTs harbouring an exon 2 duplication, a feature of many DMD patients that has not been recapitulated in culture prior to this report. We further demonstrate that this "DMD in a dish" system can be used as a pre-clinical assay to test a putative DMD therapeutic and study the mechanism of action.
Collapse
Affiliation(s)
- Majid Ebrahimi
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Heta Lad
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Aurora Fusto
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Yekaterina Tiper
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Asiman Datye
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Christine T Nguyen
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Erik Jacques
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Louise A Moyle
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Thy Nguyen
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Brennen Musgrave
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Carolina Chávez-Madero
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada
| | - Anne Bigot
- Sorbonne Universite, INSERM, Association Institut de Myologie, Centre de Recherche en Myologie, Paris UMRS974, France
| | - Chun Chen
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Scott Turner
- Pliant Therapeutics, Inc, South San Francisco, California 94080, USA
| | - Bryan A Stewart
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada; Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L1C6, Canada
| | - Elena Pegoraro
- Department of Neuroscience, University of Padua, Padua, 35128, Italy
| | - Libero Vitiello
- Department of Biology, University of Padua, Padua 35131, Italy; Interuniversity Institute of Myology (IIM), Italy
| | - Penney M Gilbert
- Donnelly Centre, University of Toronto, Toronto, ON M5S3E1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S3G9, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S3G5, Canada.
| |
Collapse
|
15
|
Yao S, Chen Z, Yu Y, Zhang N, Jiang H, Zhang G, Zhang Z, Zhang B. Current Pharmacological Strategies for Duchenne Muscular Dystrophy. Front Cell Dev Biol 2021; 9:689533. [PMID: 34490244 PMCID: PMC8417245 DOI: 10.3389/fcell.2021.689533] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked neuromuscular disorder caused by the absence of dystrophin protein, which is essential for muscle fiber integrity. Loss of dystrophin protein leads to recurrent myofiber damage, chronic inflammation, progressive fibrosis, and dysfunction of muscle stem cells. There is still no cure for DMD so far and the standard of care is principally limited to symptom relief through glucocorticoids treatments. Current therapeutic strategies could be divided into two lines. Dystrophin-targeted therapeutic strategies that aim at restoring the expression and/or function of dystrophin, including gene-based, cell-based and protein replacement therapies. The other line of therapeutic strategies aims to improve muscle function and quality by targeting the downstream pathological changes, including inflammation, fibrosis, and muscle atrophy. This review introduces the important developments in these two lines of strategies, especially those that have entered the clinical phase and/or have great potential for clinical translation. The rationale and efficacy of each agent in pre-clinical or clinical studies are presented. Furthermore, a meta-analysis of gene profiling in DMD patients has been performed to understand the molecular mechanisms of DMD.
Collapse
Affiliation(s)
- Shanshan Yao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
16
|
Swiderski K, Lynch GS. Murine models of Duchenne muscular dystrophy: is there a best model? Am J Physiol Cell Physiol 2021; 321:C409-C412. [PMID: 34260298 DOI: 10.1152/ajpcell.00212.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Kristy Swiderski
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
17
|
Lopez MA, Bontiff S, Adeyeye M, Shaibani AI, Alexander MS, Wynd S, Boriek AM. Mechanics of dystrophin deficient skeletal muscles in very young mice and effects of age. Am J Physiol Cell Physiol 2021; 321:C230-C246. [PMID: 33979214 DOI: 10.1152/ajpcell.00155.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The MDX mouse is an animal model of Duchenne muscular dystrophy, a human disease marked by an absence of the cytoskeletal protein, dystrophin. We hypothesized that 1) dystrophin serves a complex mechanical role in skeletal muscles by contributing to passive compliance, viscoelastic properties, and contractile force production and 2) age is a modulator of passive mechanics of skeletal muscles of the MDX mouse. Using an in vitro biaxial mechanical testing apparatus, we measured passive length-tension relationships in the muscle fiber direction as well as transverse to the fibers, viscoelastic stress-relaxation curves, and isometric contractile properties. To avoid confounding secondary effects of muscle necrosis, inflammation, and fibrosis, we used very young 3-wk-old mice whose muscles reflected the prefibrotic and prenecrotic state. Compared with controls, 1) muscle extensibility and compliance were greater in both along fiber direction and transverse to fiber direction in MDX mice and 2) the relaxed elastic modulus was greater in dystrophin-deficient diaphragms. Furthermore, isometric contractile muscle stress was reduced in the presence and absence of transverse fiber passive stress. We also examined the effect of age on the diaphragm length-tension relationships and found that diaphragm muscles from 9-mo-old MDX mice were significantly less compliant and less extensible than those of muscles from very young MDX mice. Our data suggest that the age of the MDX mouse is a determinant of the passive mechanics of the diaphragm; in the prefibrotic/prenecrotic stage, muscle extensibility and compliance, as well as viscoelasticity, and muscle contractility are altered by loss of dystrophin.
Collapse
Affiliation(s)
- Michael A Lopez
- Department of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, University of Alabama Birmingham, Birmingham, Alabama
| | - Sherina Bontiff
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Mary Adeyeye
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Aziz I Shaibani
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Matthew S Alexander
- Department of Pediatrics, University of Alabama Birmingham, Birmingham, Alabama
| | - Shari Wynd
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Aladin M Boriek
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
18
|
Botzenhart UU, Keil C, Tsagkari E, Zeidler-Rentzsch I, Gredes T, Gedrange T. Influence of botulinum toxin A on craniofacial morphology after injection into the right masseter muscle of dystrophin deficient (mdx-) mice. Ann Anat 2021; 236:151715. [PMID: 33675949 DOI: 10.1016/j.aanat.2021.151715] [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: 01/08/2021] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Severe craniofacial and dental abnormalities, typical for patients with progressive Duchenne muscular dystrophy (DMD), are an exellcent demonstration of Melvin L. Moss "functional matrix theory", highlighting the influence of muscle tissue on craniofacial growth and morphology. However, the currently best approved animal model for investigation of this interplay is the mdx-mouse, which offers only a limited time window for research, due to the ability of muscle regeneration, in contrast to the human course of the disease. The aim of this study was to evaluate craniofacial morphology after BTX-A induced muscle paralysis in C57Bl- and mdx-mice, to prove the suitability of BTX-A intervention to inhibit muscle regeneration in mdx-mice and thus, mimicking the human course of the DMD disease. METHODS Paralysis of the right masseter muscle was induced in 100 days old C57Bl- and mdx-mice by a single specific intramuscular BTX-A injection. Mice skulls were obtained at 21 days and 42 days after BTX-A injection and 3D radiological evaluation was performed in order to measure various craniofacial dimensions in the sagittal, transversal and vertical plane. Statstical analysis were performed using SigmaStat®Version 3.5. In case of normal distribution, unpaired t-test and otherwise the Mann-Whitney-U test was applied. A statistical significance was given in case of p ≤ 0.05. RESULTS In contrast to C57Bl-mice, in mdx-mice, three weeks after BTX-A treatment a significant decrease of skull dimensions was noted in most of the measurements followed by a significant increase at the second investigation period. CONCLUSIONS BTX-A can induce changes in craniofacial morphology and presumably partially inhibit muscle regeneration in mdx-mice, but cannot completely intensify craniofacial effects elicited by dystrophy. Further research is necessary in order to fully understand muscle-bone interplay after BTX-A injection into dystrophic muscles.
Collapse
Affiliation(s)
| | - Christiane Keil
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany; Department of Orthodontics, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Eirini Tsagkari
- Department of Orthodontics, Faculty of Dentistry School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Ines Zeidler-Rentzsch
- Department of Otorhinolaryngology, Head and Neck Surgery, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Tomasz Gredes
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany; Department of Orthodontics, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Tomasz Gedrange
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| |
Collapse
|
19
|
Brolin C, Lim EWK, Nielsen PE. In Vivo Administration of Splice Switching PNAs Using the mdx Mouse as a Model System. Methods Mol Biol 2021; 2105:241-250. [PMID: 32088875 DOI: 10.1007/978-1-0716-0243-0_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common and severe form of muscular dystrophy and is caused by gene mutations that abolish production of functional dystrophin muscle protein. A promising new treatment exploits specifically targeted RNA-acting drugs that are able to partially restore the dystrophin protein. The mdx mouse model (animal model of DMD) serves as a good in vivo model for testing these antisense drugs. The simplest in vivo test, which circumvents the systemic circulation, is intramuscular administration of the compound. After 7 days it is possible to detect exon skipping by reverse transcriptase PCR, and newly synthesized dystrophin-positive fibers by immunohistochemistry and western blotting. All muscles, including the heart, are affected by the disease and must be treated. Therefore the use of antisense therapy for treatment of DMD requires systemic administration, and the model is also useful for systemic administration.
Collapse
Affiliation(s)
- Camilla Brolin
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ernest Wee Kiat Lim
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter E Nielsen
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
20
|
Kröger S, Watkins B. Muscle spindle function in healthy and diseased muscle. Skelet Muscle 2021; 11:3. [PMID: 33407830 PMCID: PMC7788844 DOI: 10.1186/s13395-020-00258-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/20/2020] [Indexed: 12/16/2022] Open
Abstract
Almost every muscle contains muscle spindles. These delicate sensory receptors inform the central nervous system (CNS) about changes in the length of individual muscles and the speed of stretching. With this information, the CNS computes the position and movement of our extremities in space, which is a requirement for motor control, for maintaining posture and for a stable gait. Many neuromuscular diseases affect muscle spindle function contributing, among others, to an unstable gait, frequent falls and ataxic behavior in the affected patients. Nevertheless, muscle spindles are usually ignored during examination and analysis of muscle function and when designing therapeutic strategies for neuromuscular diseases. This review summarizes the development and function of muscle spindles and the changes observed under pathological conditions, in particular in the various forms of muscular dystrophies.
Collapse
Affiliation(s)
- Stephan Kröger
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152, Planegg-Martinsried, Germany.
| | - Bridgette Watkins
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Großhaderner Str. 9, 82152, Planegg-Martinsried, Germany
| |
Collapse
|
21
|
Taetzsch T, Shapiro D, Eldosougi R, Myers T, Settlage RE, Valdez G. The microRNA miR-133b functions to slow Duchenne muscular dystrophy pathogenesis. J Physiol 2021; 599:171-192. [PMID: 32991751 PMCID: PMC8418193 DOI: 10.1113/jp280405] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/14/2020] [Indexed: 01/22/2023] Open
Abstract
KEY POINTS Impairment of muscle biogenesis contributes to the progression of Duchenne muscular dystrophy (DMD). As a muscle enriched microRNA that has been implicated in muscle biogenesis, the role of miR-133b in DMD remains unknown. To assess miR-133b function in DMD-affected skeletal muscles, we genetically ablated miR-133b in the mdx mouse model of DMD. We show that deletion of miR-133b exacerbates the dystrophic phenotype of DMD-afflicted skeletal muscle by dysregulating muscle stem cells involved in muscle biogenesis, in addition to affecting signalling pathways related to inflammation and fibrosis. Our results provide evidence that miR-133b may underlie DMD pathology by affecting the proliferation and differentiation of muscle stem cells. ABSTRACT Duchenne muscular dystrophy (DMD) is characterized by progressive skeletal muscle degeneration. No treatments are currently available to prevent the disease. While the muscle enriched microRNA miR-133b has been implicated in muscle biogenesis, its role in DMD remains unknown. To assess miR-133b function in DMD-affected skeletal muscles, we genetically ablated miR-133b in the mdx mouse model of DMD. In the absence of miR-133b, the tibialis anterior muscle of P30 mdx mice is smaller in size and exhibits a thickened interstitial space containing more mononucleated cells. Additional analysis revealed that miR-133b deletion influences muscle fibre regeneration, satellite cell proliferation and differentiation, and induces widespread transcriptomic changes in mdx muscle. These include known miR-133b targets as well as genes involved in cell proliferation and fibrosis. Altogether, our data demonstrate that skeletal muscles utilize miR-133b to mitigate the deleterious effects of DMD.
Collapse
Affiliation(s)
- Thomas Taetzsch
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Dillon Shapiro
- Molecular Biology, Cell Biology, & Biochemistry Graduate Program, Brown University, Providence, RI, USA
| | - Randa Eldosougi
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | - Tracey Myers
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, USA
| | | | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, United States
| |
Collapse
|
22
|
Rybalka E, Timpani CA, Debruin DA, Bagaric RM, Campelj DG, Hayes A. The Failed Clinical Story of Myostatin Inhibitors against Duchenne Muscular Dystrophy: Exploring the Biology behind the Battle. Cells 2020; 9:E2657. [PMID: 33322031 PMCID: PMC7764137 DOI: 10.3390/cells9122657] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
Myostatin inhibition therapy has held much promise for the treatment of muscle wasting disorders. This is particularly true for the fatal myopathy, Duchenne Muscular Dystrophy (DMD). Following on from promising pre-clinical data in dystrophin-deficient mice and dogs, several clinical trials were initiated in DMD patients using different modality myostatin inhibition therapies. All failed to show modification of disease course as dictated by the primary and secondary outcome measures selected: the myostatin inhibition story, thus far, is a failed clinical story. These trials have recently been extensively reviewed and reasons why pre-clinical data collected in animal models have failed to translate into clinical benefit to patients have been purported. However, the biological mechanisms underlying translational failure need to be examined to ensure future myostatin inhibitor development endeavors do not meet with the same fate. Here, we explore the biology which could explain the failed translation of myostatin inhibitors in the treatment of DMD.
Collapse
Affiliation(s)
- Emma Rybalka
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Cara A. Timpani
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Danielle A. Debruin
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Ryan M. Bagaric
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Dean G. Campelj
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
| | - Alan Hayes
- Institute for Health and Sport (IHeS), Victoria University, Melbourne, Victoria 8001, Australia; (D.A.D.); (R.M.B.); (D.G.C.); (A.H.)
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, St Albans, Victoria 3021, Australia
- Department of Medicine—Western Health, Melbourne Medical School, The University of Melbourne, Melbourne, 3021 Victoria, Australia
| |
Collapse
|
23
|
Baumann CW, Warren GL, Lowe DA. Plasmalemma Function Is Rapidly Restored in Mdx Muscle after Eccentric Contractions. Med Sci Sports Exerc 2020; 52:354-361. [PMID: 31415447 DOI: 10.1249/mss.0000000000002126] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE Muscle that lacks dystrophin, as in the mdx mouse, has a heightened sensitivity to eccentric (ECC) contraction-induced strength loss but an enhanced rate of recovery. However, the timeline and mechanisms underlying why mdx muscle recovers quicker have yet to be determined. We used an EMG approach to analyze plasmalemma electrophysiological function during and after ECC contraction-induced injury to test the hypothesis that loss of plasmalemmal excitability is a transient event in mdx muscle. METHODS Mice were implanted with stimulating electrodes on the common peroneal nerve and EMG electrodes on the tibialis anterior muscle. Anterior crural muscles of anesthetized mice performed one or two bouts of 50 injurious ECC contractions, and recovery of maximal isometric torque and M-wave root mean square (RMS) were assessed after each bout. RESULTS Maximal isometric torque and M-wave RMS were equally reduced 62% (P < 0.001) in mdx mice immediately after the initial ECC injury. For these mdx mice, M-wave RMS was still reduced at 2 d postinjury (P = 0.034) but was not different from preinjury values by 6 d (P = 0.106), whereas torque took up to 9 d to recover (P = 0.333). M-wave RMS did not change (P = 0.390) in wild-type mice in response to ECC injury, whereas torque decreased 35% (P < 0.001) and recovered by day 2 (P = 0.311). Results from the second bout of ECC contractions were similar to those observed during and after the initial injury. CONCLUSION Functional dystrophin is necessary for excitation to occur at the plasmalemma during ECC contractions but is not essential for the complete recovery of plasmalemma electrophysiological function or maximal isometric strength.
Collapse
Affiliation(s)
- Cory W Baumann
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, MN
| | - Gordon L Warren
- Department of Physical Therapy, Georgia State University, Atlanta, GA
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, Medical School, University of Minnesota, Minneapolis, MN
| |
Collapse
|
24
|
Krishnan VS, Thanigaiarasu LP, White R, Crew R, Larcher T, Le Guiner C, Grounds MD. Dystrophic Dmd mdx rats show early neuronal changes (increased S100β and Tau5) at 8 months, supporting severe dystropathology in this rodent model of Duchenne muscular dystrophy. Mol Cell Neurosci 2020; 108:103549. [PMID: 32890728 DOI: 10.1016/j.mcn.2020.103549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/25/2022] Open
Abstract
The intrinsic necrosis of skeletal muscles in animal models of Duchenne muscular dystrophy (DMD) damages neuromuscular junctions (NMJs) with progressively altered NMJs associated with denervation and premature changes in dystrophic nerves. In the mdx mouse model of DMD, the proteins S100β and Tau5 are significantly increased in sciatic nerves by 13 months (M) of age, far earlier (by 9 M) than in normal wildtype (WT) nerves. Since dystrophic Dmdmdx rats are reported to have a more severe dystropathology than mdx mice, we hypothesised that Dmdmdx rat nerves would show earlier neuronal changes compared with mdx nerves. We quantified levels of 8 proteins (by immunoblotting) in sciatic and radial nerves from young adult Dmdmdx rats (aged 8 M) and mdx mice (9 M), plus levels of 7 mRNAs (by qPCR) in rat nerves only. Sciatic nerves of 8 M Dmdmdx rats had more consistently increased levels of S100β and Tau5 proteins, compared with 9 M mdx mice, supporting pronounced dystropathology in the rat model. There were no differences for mRNA levels, apart from higher gelsolin mRNA in Dmdmdx sciatic nerves. The pronounced protein changes in Dmdmdx nerves indicate a severe ongoing myonecrosis, and likely consequent myofibre denervation, for the dystrophic rat model. These data support increased neuronal proteins in dystrophic nerves as a novel pre-clinical readout of ongoing myonecrosis for DMD research. In older DMD boys, such progressive neuronal changes over many years are likely to contribute to loss of muscle function, and may complicate evaluation of late-onset clinical therapies.
Collapse
Affiliation(s)
- Vidya S Krishnan
- School of Human Sciences, the University of Western Australia, Australia, 6009
| | | | - Robert White
- School of Human Sciences, the University of Western Australia, Australia, 6009
| | - Rachael Crew
- School of Human Sciences, the University of Western Australia, Australia, 6009
| | | | - Caroline Le Guiner
- INSERM UMR1089, University of Nantes, Translational Research for Neuromuscular Diseases, Nantes, France
| | - Miranda D Grounds
- School of Human Sciences, the University of Western Australia, Australia, 6009.
| |
Collapse
|
25
|
Persiconi I, Cosmi F, Guadagno NA, Lupo G, De Stefano ME. Dystrophin Is Required for the Proper Timing in Retinal Histogenesis: A Thorough Investigation on the mdx Mouse Model of Duchenne Muscular Dystrophy. Front Neurosci 2020; 14:760. [PMID: 32982660 PMCID: PMC7487415 DOI: 10.3389/fnins.2020.00760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked muscular disease caused by defective expression of the cytoskeletal protein dystrophin (Dp427). Selected autonomic and central neurons, including retinal neurons, express Dp427 and/or dystrophin shorter isoforms. Because of this, DMD patients may also experience different forms of cognitive impairment, neurological and autonomic disorders, and specific visual defects. DMD-related damages to the nervous system are established during development, suggesting a role for all dystrophin isoforms in neural circuit development and differentiation; however, to date, their function in retinogenesis has never been investigated. In this large-scale study, we analyzed whether the lack of Dp427 affects late retinogenesis in the mdx mouse, the most well studied animal model of DMD. Retinal gene expression and layer maturation, as well as neural cell proliferation, apoptosis, and differentiation, were evaluated in E18 and/or P0, P5, P10, and adult mice. In mdx mice, expression of Capn3, Id3 (E18-P5), and Dtnb (P5) genes, encoding proteins involved in different aspects of retina development and synaptogenesis (e.g., Calpain 3, DNA-binding protein inhibitor-3, and β-dystrobrevin, respectively), was transiently reduced compared to age-matched wild type mice. Concomitantly, a difference in the time required for the retinal ganglion cell layer to reach appropriate thickness was observed (P0–P5). Immunolabeling for specific cell markers also evidenced a significant dysregulation in the number of GABAergic amacrine cells (P5–P10), a transient decrease in the area immunopositive for the Vesicular Glutamate Transporter 1 (VGluT1) during ribbon synapse maturation (P10) and a reduction in the number of calretinin+ retinal ganglion cells (RGCs) (adults). Finally, the number of proliferating retinal progenitor cells (P5–P10) and apoptotic cells (P10) was reduced. These results support the hypothesis of a role for Dp427 during late retinogenesis different from those proposed in consolidated neural circuits. In particular, Dp427 may be involved in shaping specific steps of retina differentiation. Notably, although most of the above described quantitative alterations recover over time, the number of calretinin+ RGCs is reduced only in the mature retina. This suggests that alterations subtler than the timing of retinal maturation may occur, a hypothesis that demands further in-depth functional studies.
Collapse
Affiliation(s)
- Irene Persiconi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Francesca Cosmi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | | | - Giuseppe Lupo
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.,Center for Research in Neurobiology "Daniel Bovet", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
26
|
Barthélémy I, Calmels N, Weiss RB, Tiret L, Vulin A, Wein N, Peccate C, Drougard C, Beroud C, Deburgrave N, Thibaud JL, Escriou C, Punzón I, Garcia L, Kaplan JC, Flanigan KM, Leturcq F, Blot S. X-linked muscular dystrophy in a Labrador Retriever strain: phenotypic and molecular characterisation. Skelet Muscle 2020; 10:23. [PMID: 32767978 PMCID: PMC7412789 DOI: 10.1186/s13395-020-00239-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background Canine models of Duchenne muscular dystrophy (DMD) are a valuable tool to evaluate potential therapies because they faithfully reproduce the human disease. Several cases of dystrophinopathies have been described in canines, but the Golden Retriever muscular dystrophy (GRMD) model remains the most used in preclinical studies. Here, we report a new spontaneous dystrophinopathy in a Labrador Retriever strain, named Labrador Retriever muscular dystrophy (LRMD). Methods A colony of LRMD dogs was established from spontaneous cases. Fourteen LRMD dogs were followed-up and compared to the GRMD standard using several functional tests. The disease causing mutation was studied by several molecular techniques and identified using RNA-sequencing. Results The main clinical features of the GRMD disease were found in LRMD dogs; the functional tests provided data roughly overlapping with those measured in GRMD dogs, with similar inter-individual heterogeneity. The LRMD causal mutation was shown to be a 2.2-Mb inversion disrupting the DMD gene within intron 20 and involving the TMEM47 gene. In skeletal muscle, the Dp71 isoform was ectopically expressed, probably as a consequence of the mutation. We found no evidence of polymorphism in either of the two described modifier genes LTBP4 and Jagged1. No differences were found in Pitpna mRNA expression levels that would explain the inter-individual variability. Conclusions This study provides a full comparative description of a new spontaneous canine model of dystrophinopathy, found to be phenotypically equivalent to the GRMD model. We report a novel large DNA mutation within the DMD gene and provide evidence that LRMD is a relevant model to pinpoint additional DMD modifier genes.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Nadège Calmels
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Laboratoire de Diagnostic Génétique-Institut de Génétique Médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, 1 Place de L'Hôpital, 67091, Strasbourg, France
| | - Robert B Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Adeline Vulin
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France
| | - Nicolas Wein
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Cécile Peccate
- SQY Therapeutics, Université de Versailles Saint-Quentin-en-Yvelines, Montigny le Bretonneux, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Carole Drougard
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Christophe Beroud
- Aix Marseille Université, INSERM, MMG, Bioinformatics & Genetics, Marseille, France.,APHM, Hôpital Timone Enfants, Laboratoire de Génétique Moléculaire, Marseille, France
| | - Nathalie Deburgrave
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Jean-Laurent Thibaud
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Catherine Escriou
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Isabel Punzón
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Luis Garcia
- Université de Versailles Saint-Quentin-en-Yvelines, U1179 INSERM, UFR des Sciences de la Santé, Montigny le Bretonneux, France
| | - Jean-Claude Kaplan
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France
| | - Kevin M Flanigan
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - France Leturcq
- Laboratoire de biochimie et génétique moléculaire, hôpital Cochin, AP-HP, université Paris Descartes-Sorbonne Paris Cité, Paris, France.,Sorbonne Universités, UPMC Université Paris 06, INSERM UMRS974, Centre de Recherche en Myologie, Institut de Myologie, G.H. Pitié Salpêtrière, Paris, France
| | - Stéphane Blot
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France.
| |
Collapse
|
27
|
Boccanegra B, Verhaart IEC, Cappellari O, Vroom E, De Luca A. Safety issues and harmful pharmacological interactions of nutritional supplements in Duchenne muscular dystrophy: considerations for Standard of Care and emerging virus outbreaks. Pharmacol Res 2020; 158:104917. [PMID: 32485610 PMCID: PMC7261230 DOI: 10.1016/j.phrs.2020.104917] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 05/08/2020] [Accepted: 05/08/2020] [Indexed: 12/13/2022]
Abstract
At the moment, little treatment options are available for Duchenne muscular dystrophy (DMD). The absence of the dystrophin protein leads to a complex cascade of pathogenic events in myofibres, including chronic inflammation and oxidative stress as well as altered metabolism. The attention towards dietary supplements in DMD is rapidly increasing, with the aim to counteract pathology-related alteration in nutrient intake, the consequences of catabolic distress or to enhance the immunological response of patients as nowadays for the COVID-19 pandemic emergency. By definition, supplements do not exert therapeutic actions, although a great confusion may arise in daily life by the improper distinction between supplements and therapeutic compounds. For most supplements, little research has been done and little evidence is available concerning their effects in DMD as well as their preventing actions against infections. Often these are not prescribed by clinicians and patients/caregivers do not discuss the use with their clinical team. Then, little is known about the real extent of supplement use in DMD patients. It is mistakenly assumed that, since compounds are of natural origin, if a supplement is not effective, it will also do no harm. However, supplements can have serious side effects and also have harmful interactions, in terms of reducing efficacy or leading to toxicity, with other therapies. It is therefore pivotal to shed light on this unclear scenario for the sake of patients. This review discusses the supplements mostly used by DMD patients, focusing on their potential toxicity, due to a variety of mechanisms including pharmacodynamic or pharmacokinetic interactions and contaminations, as well as on reports of adverse events. This overview underlines the need for caution in uncontrolled use of dietary supplements in fragile populations such as DMD patients. A culture of appropriate use has to be implemented between clinicians and patients' groups.
Collapse
Affiliation(s)
- Brigida Boccanegra
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Ingrid E C Verhaart
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands; Duchenne Parent Project, the Netherlands
| | - Ornella Cappellari
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Elizabeth Vroom
- Duchenne Parent Project, the Netherlands; World Duchenne Organisation (UPPMD), the Netherlands
| | - Annamaria De Luca
- Unit of Pharmacology, Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
28
|
Barthélémy I, Hitte C, Tiret L. The Dog Model in the Spotlight: Legacy of a Trustful Cooperation. J Neuromuscul Dis 2020; 6:421-451. [PMID: 31450509 PMCID: PMC6918919 DOI: 10.3233/jnd-190394] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Dogs have long been used as a biomedical model system and in particular as a preclinical proof of concept for innovative therapies before translation to humans. A recent example of the utility of this animal model is the promising myotubularin gene delivery in boys affected by X-linked centronuclear myopathy after successful systemic, long-term efficient gene therapy in Labrador retrievers. Mostly, this is due to unique features that make dogs an optimal system. The continuous emergence of spontaneous inherited disorders enables the identification of reliable complementary molecular models for human neuromuscular disorders (NMDs). Dogs’ characteristics including size, lifespan and unprecedented medical care level allow a comprehensive longitudinal description of diseases. Moreover, the highly similar pathogenic mechanisms with human patients yield to translational robustness. Finally, interindividual phenotypic heterogeneity between dogs helps identifying modifiers and anticipates precision medicine issues. This review article summarizes the present list of molecularly characterized dog models for NMDs and provides an exhaustive list of the clinical and paraclinical assays that have been developed. This toolbox offers scientists a sensitive and reliable system to thoroughly evaluate neuromuscular function, as well as efficiency and safety of innovative therapies targeting these NMDs. This review also contextualizes the model by highlighting its unique genetic value, shaped by the long-term coevolution of humans and domesticated dogs. Because the dog is one of the most protected research animal models, there is considerable opposition to include it in preclinical projects, posing a threat to the use of this model. We thus discuss ethical issues, emphasizing that unlike many other models, the dog also benefits from its contribution to comparative biomedical research with a drastic reduction in the prevalence of morbid alleles in the breeding stock and an improvement in medical care.
Collapse
Affiliation(s)
- Inès Barthélémy
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| | - Christophe Hitte
- CNRS, University of Rennes 1, UMR 6290, IGDR, Faculty of Medicine, SFR Biosit, Rennes, France
| | - Laurent Tiret
- U955 - IMRB, Team 10 - Biology of the neuromuscular system, Inserm, UPEC, EFS, École nationale vétérinaire d'Alfort, Maisons-Alfort, France
| |
Collapse
|
29
|
Lim KRQ, Nguyen Q, Dzierlega K, Huang Y, Yokota T. CRISPR-Generated Animal Models of Duchenne Muscular Dystrophy. Genes (Basel) 2020; 11:genes11030342. [PMID: 32213923 PMCID: PMC7141101 DOI: 10.3390/genes11030342] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked recessive neuromuscular disorder most commonly caused by mutations disrupting the reading frame of the dystrophin (DMD) gene. DMD codes for dystrophin, which is critical for maintaining the integrity of muscle cell membranes. Without dystrophin, muscle cells receive heightened mechanical stress, becoming more susceptible to damage. An active body of research continues to explore therapeutic treatments for DMD as well as to further our understanding of the disease. These efforts rely on having reliable animal models that accurately recapitulate disease presentation in humans. While current animal models of DMD have served this purpose well to some extent, each has its own limitations. To help overcome this, clustered regularly interspaced short palindromic repeat (CRISPR)-based technology has been extremely useful in creating novel animal models for DMD. This review focuses on animal models developed for DMD that have been created using CRISPR, their advantages and disadvantages as well as their applications in the DMD field.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Kasia Dzierlega
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Yiqing Huang
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (K.R.Q.L.); (Q.N.); (K.D.); (Y.H.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G 2H7, Canada
- Correspondence: ; Tel.: +1-780-492-1102
| |
Collapse
|
30
|
van Putten M, Lloyd EM, de Greef JC, Raz V, Willmann R, Grounds MD. Mouse models for muscular dystrophies: an overview. Dis Model Mech 2020; 13:dmm043562. [PMID: 32224495 PMCID: PMC7044454 DOI: 10.1242/dmm.043562] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Muscular dystrophies (MDs) encompass a wide variety of inherited disorders that are characterized by loss of muscle tissue associated with a progressive reduction in muscle function. With a cure lacking for MDs, preclinical developments of therapeutic approaches depend on well-characterized animal models that recapitulate the specific pathology in patients. The mouse is the most widely and extensively used model for MDs, and it has played a key role in our understanding of the molecular mechanisms underlying MD pathogenesis. This has enabled the development of therapeutic strategies. Owing to advancements in genetic engineering, a wide variety of mouse models are available for the majority of MDs. Here, we summarize the characteristics of the most commonly used mouse models for a subset of highly studied MDs, collated into a table. Together with references to key publications describing these models, this brief but detailed overview would be useful for those interested in, or working with, mouse models of MD.
Collapse
Affiliation(s)
- Maaike van Putten
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Erin M Lloyd
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| | - Jessica C de Greef
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Vered Raz
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | | | - Miranda D Grounds
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| |
Collapse
|
31
|
Gerwin L, Rossmanith S, Haupt C, Schultheiß J, Brinkmeier H, Bittner RE, Kröger S. Impaired muscle spindle function in murine models of muscular dystrophy. J Physiol 2020; 598:1591-1609. [PMID: 32003874 DOI: 10.1113/jp278563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Muscular dystrophy patients suffer from progressive degeneration of skeletal muscle fibres, sudden spontaneous falls, balance problems, as well as gait and posture abnormalities. Dystrophin- and dysferlin-deficient mice, models for different types of muscular dystrophy with different aetiology and molecular basis, were characterized to investigate if muscle spindle structure and function are impaired. The number and morphology of muscle spindles were unaltered in both dystrophic mouse lines but muscle spindle resting discharge and their responses to stretch were altered. In dystrophin-deficient muscle spindles, the expression of the paralogue utrophin was substantially upregulated, potentially compensating for the dystrophin deficiency. The results suggest that muscle spindles might contribute to the motor problems observed in patients with muscular dystrophy. ABSTRACT Muscular dystrophies comprise a heterogeneous group of hereditary diseases characterized by progressive degeneration of extrafusal muscle fibres as well as unstable gait and frequent falls. To investigate if muscle spindle function is impaired, we analysed their number, morphology and function in wildtype mice and in murine model systems for two distinct types of muscular dystrophy with very different disease aetiology, i.e. dystrophin- and dysferlin-deficient mice. The total number and the overall structure of muscle spindles in soleus muscles of both dystrophic mouse mutants appeared unchanged. Immunohistochemical analyses of wildtype muscle spindles revealed a concentration of dystrophin and β-dystroglycan in intrafusal fibres outside the region of contact with the sensory neuron. While utrophin was absent from the central part of intrafusal fibres of wildtype mice, it was substantially upregulated in dystrophin-deficient mice. Single-unit extracellular recordings of sensory afferents from muscle spindles of the extensor digitorum longus muscle revealed that muscle spindles from both dystrophic mouse strains have an increased resting discharge and a higher action potential firing rate during sinusoidal vibrations, particularly at low frequencies. The response to ramp-and-hold stretches appeared unaltered compared to the respective wildtype mice. We observed no exacerbated functional changes in dystrophin and dysferlin double mutant mice compared to the single mutant animals. These results show alterations in muscle spindle afferent responses in both dystrophic mouse lines, which might cause an increased muscle tone, and might contribute to the unstable gait and frequent falls observed in patients with muscular dystrophy.
Collapse
Affiliation(s)
- Laura Gerwin
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Großhaderner Str. 9, D-82152, Planegg-Martinsried, Germany.,Institute for Stem Cell Research, German Research Center for Environmental Health, Helmholtz Centre Munich, Ingolstädter Landstraße 1, D-85764, Neuherberg, Germany
| | - Sarah Rossmanith
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Großhaderner Str. 9, D-82152, Planegg-Martinsried, Germany
| | - Corinna Haupt
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Großhaderner Str. 9, D-82152, Planegg-Martinsried, Germany
| | - Jürgen Schultheiß
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Großhaderner Str. 9, D-82152, Planegg-Martinsried, Germany
| | - Heinrich Brinkmeier
- Institute for Pathophysiology, University Medicine Greifswald, Martin-Luther-Str. 6, 17489, Greifswald, Germany
| | - Reginald E Bittner
- Neuromuscular Research Department, Center for Anatomy and Cell Biology, Medical University of Vienna, Waehringerstrasse 13, 1090, Vienna, Austria
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University, Großhaderner Str. 9, D-82152, Planegg-Martinsried, Germany
| |
Collapse
|
32
|
Dickson PE, Mittleman G. Visual Discrimination, Serial Reversal, and Extinction Learning in the mdx Mouse. Front Behav Neurosci 2019; 13:200. [PMID: 31543764 PMCID: PMC6728792 DOI: 10.3389/fnbeh.2019.00200] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy and the most common neuromuscular disorder. In addition to neuromuscular consequences, some individuals with DMD experience global intellectual dysfunction and executive dysfunction of unknown mechanistic origin. The cognitive profile of the mdx mouse, the most commonly used mouse model of DMD, has been incompletely characterized and has never been assessed using the touchscreen operant conditioning paradigm. The touchscreen paradigm allows the use of protocols that are virtually identical to those used in human cognitive testing and may, therefore, provide the most translational paradigm for quantifying mouse cognitive function. In the present study, we used the touchscreen paradigm to assess the effects of the mdx mutation on visual discrimination learning, serial reversal learning, and extinction learning. To enable measuring task-dependent learning and memory processes while holding demands on sensory-driven information processing constant, we developed equally salient visual stimuli and used them on all experimental stages. Acquisition of the initial pairwise visual discrimination was facilitated in mdx mice relative to wildtype littermates; this effect was not explained by genotypic differences in impulsivity, motivation, or motor deficits. The mdx mutation had no effect on serial reversal or extinction learning. Together, findings from this study and previous studies suggest that mdx effects on cognitive function are task-specific and may be influenced by discrimination type (spatial, visual), reward type (food, escape from a non-preferred environment), sex, and genetic background.
Collapse
Affiliation(s)
| | - Guy Mittleman
- Department of Psychological Science, Ball State University, Muncie, IN, United States
| |
Collapse
|
33
|
Verhaart IEC, Putker K, van de Vijver D, Tanganyika-de Winter CL, Pasteuning-Vuhman S, Plomp JJ, Aartsma-Rus AM, van Putten M. Cross-sectional study into age-related pathology of mouse models for limb girdle muscular dystrophy types 2D and 2F. PLoS One 2019; 14:e0220665. [PMID: 31430305 PMCID: PMC6701749 DOI: 10.1371/journal.pone.0220665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Limb girdle muscular dystrophy (LGMD) types 2D and 2F are caused by mutations in the genes encoding for α- and δ-sarcoglycan, respectively, leading to progressive muscle weakness. Mouse models exist for LGMD2D (Sgca-/-) and 2F (Sgcd-/-). In a previous natural history study, we described the pathology in these mice at 34 weeks of age. However, the development of muscle pathology at younger ages has not been fully characterised yet. We therefore performed a study into age-related changes in muscle function and pathology by examining mice at different ages. From 4 weeks of age onwards, male mice were subjected to functional tests and sacrificed at respectively 8, 16 or 24 weeks of age. Muscle histopathology and expression of genes involved in muscle pathology were analysed for several skeletal muscles, while miRNA levels were assessed in serum. In addition, for Sgcd-/- mice heart pathology was assessed. Muscle function showed a gradual decline in both Sgca-/- and Sgcd-/- mice. Respiratory function was also impaired at all examined timepoints. Already at 8 weeks of age, muscle pathology was prominent, and fibrotic, inflammatory and regenerative markers were elevated, which remained relatively constant with age. In addition, Sgcd-/- mice showed signs of cardiomyopathy from 16 weeks of age onwards. These results indicate that Sgca-/- and Sgcd-/- are relevant disease models for LGMD2D and 2F.
Collapse
Affiliation(s)
- Ingrid E. C. Verhaart
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Kayleigh Putker
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Davy van de Vijver
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Jaap J. Plomp
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annemieke M. Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
34
|
Xu R, Jia Y, Zygmunt DA, Martin PT. rAAVrh74.MCK.GALGT2 Protects against Loss of Hemodynamic Function in the Aging mdx Mouse Heart. Mol Ther 2019; 27:636-649. [PMID: 30711447 PMCID: PMC6403484 DOI: 10.1016/j.ymthe.2019.01.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 01/16/2023] Open
Abstract
Dilated cardiomyopathy is a common cause of death in patients with Duchenne muscular dystrophy (DMD). Gene therapies for DMD must, therefore, have a therapeutic impact in cardiac as well as skeletal muscles. Our previous studies have shown that GALGT2 overexpression in mdx skeletal muscles can prevent muscle damage. Here we have tested whether rAAVrh74.MCK.GALGT2 gene therapy in mdx cardiac muscle can prevent the loss of heart function. Treatment of mdx hearts with rAAVrh74.MCK.GALGT2 1 day after birth did not negatively alter hemodynamic function, tested at 3 months of age, and it prevented early left ventricular remodeling and expression of fibrotic gene markers. Intravenous treatment of mdx mice with rAAVrh74.MCK.GALGT2 at 2 months of age significantly improved stroke volume and cardiac output compared to mock-treated mice analyzed at 17 months, both at rest and after stimulation with dobutamine. rAAVrh74.MCK.GALGT2 treatment of mdx heart correlated with increased glycosylation of α-dystroglycan with the CT glycan and increased utrophin protein expression. These data provide the first demonstration that GALGT2 overexpression can inhibit the loss of cardiac function in the dystrophin-deficient heart and, thus, may benefit both cardiac and skeletal muscles in DMD patients.
Collapse
Affiliation(s)
- Rui Xu
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Ying Jia
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
35
|
Truskey GA. Development and application of human skeletal muscle microphysiological systems. LAB ON A CHIP 2018; 18:3061-3073. [PMID: 30183050 PMCID: PMC6177290 DOI: 10.1039/c8lc00553b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
A number of major disease states involve skeletal muscle, including type 2 diabetes, muscular dystrophy, sarcopenia and cachexia arising from cancer or heart disease. Animals do not accurately represent many of these disease states. Human skeletal muscle microphysiological systems derived from primary or induced pluripotent stem cells (hPSCs) can provide an in vitro model of genetic and chronic diseases and assess individual variations. Three-dimensional culture systems more accurately represent skeletal muscle function than do two-dimensional cultures. While muscle biopsies enable culture of primary muscle cells, hPSCs provide the opportunity to sample a wider population of donors. Recent advances to promote maturation of PSC-derived skeletal muscle provide an alternative to primary cells. While contractile function is often measured in three-dimensional cultures and several systems exist to characterize contraction of small numbers of muscle fibers, there is a need for functional measures of metabolism suited for microphysiological systems. Future research should address generation of well-differentiated hPSC-derived muscle cells, enabling muscle repair in vitro, and improved disease models.
Collapse
Affiliation(s)
- George A Truskey
- Department of Biomedical Engineering, Duke University, 1427 CIEMAS, 101 Science Drive, Durham, NC 27708-0281, USA.
| |
Collapse
|
36
|
Taurine and Methylprednisolone Administration at Close Proximity to the Onset of Muscle Degeneration Is Ineffective at Attenuating Force Loss in the Hind-Limb of 28 Days Mdx Mice. Sports (Basel) 2018; 6:sports6040109. [PMID: 30274388 PMCID: PMC6315906 DOI: 10.3390/sports6040109] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 01/01/2023] Open
Abstract
An increasing number of studies have shown supplementation with the amino acid taurine to have promise in ameliorating dystrophic symptoms in the mdx mouse model of Duchenne Muscular Dystrophy (DMD). Here we build on this limited body of work by investigating the efficacy of supplementing mdx mice with taurine postnatally at a time suggestive of when dystrophic symptoms would begin to manifest in humans, and when treatments would likely begin. Mdx mice were given either taurine (mdx tau), the steroid alpha methylprednisolone (PDN), or tau + PDN (mdx tau + PDN). Taurine (2.5% wt/vol) enriched drinking water was given from 14 days and PDN (1 mg/kg daily) from 18 days. Wild-type (WT, C57BL10/ScSn) mice were used as a control to mdx mice to represent healthy tissue. In the mdx mouse, peak damage occurs at 28 days, and in situ assessment of contractile characteristics showed that taurine, PDN, and the combined taurine + PDN treatment was ineffective at attenuating the force loss experienced by mdx mice. Given the benefits of taurine as well as methylprednisolone reported previously, when supplemented at close proximity to the onset of severity muscle degeneration these benefits are no longer apparent.
Collapse
|
37
|
Valladares D, Utreras-Mendoza Y, Campos C, Morales C, Diaz-Vegas A, Contreras-Ferrat A, Westermeier F, Jaimovich E, Marchi S, Pinton P, Lavandero S. IP 3 receptor blockade restores autophagy and mitochondrial function in skeletal muscle fibers of dystrophic mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3685-3695. [PMID: 30251688 DOI: 10.1016/j.bbadis.2018.08.042] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/06/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022]
Abstract
Duchenne muscular dystrophy (DMD) is characterized by a severe and progressive destruction of muscle fibers associated with altered Ca2+ homeostasis. We have previously shown that the IP3 receptor (IP3R) plays a role in elevating basal cytoplasmic Ca2+ and that pharmacological blockade of IP3R restores muscle function. Moreover, we have shown that the IP3R pathway negatively regulates autophagy by controlling mitochondrial Ca2+ levels. Nevertheless, it remains unclear whether IP3R is involved in abnormal mitochondrial Ca2+ levels, mitochondrial dynamics, or autophagy and mitophagy observed in adult DMD skeletal muscle. Here, we show that the elevated basal autophagy and autophagic flux levels were normalized when IP3R was downregulated in mdx fibers. Pharmacological blockade of IP3R in mdx fibers restored both increased mitochondrial Ca2+ levels and mitochondrial membrane potential under resting conditions. Interestingly, mdx mitochondria changed from a fission to an elongated state after IP3R knockdown, and the elevated mitophagy levels in mdx fibers were normalized. To our knowledge, this is the first study associating IP3R1 activity with changes in autophagy, mitochondrial Ca2+ levels, mitochondrial membrane potential, mitochondrial dynamics, and mitophagy in adult mouse skeletal muscle. Moreover, these results suggest that increased IP3R activity in mdx fibers plays an important role in the pathophysiology of DMD. Overall, these results lead us to propose the use of specific IP3R blockers as a new pharmacological treatment for DMD, given their ability to restore both autophagy/mitophagy and mitochondrial function.
Collapse
Affiliation(s)
- Denisse Valladares
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Escuela de Kinesiologia, Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile.
| | - Yildy Utreras-Mendoza
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Cristian Campos
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Camilo Morales
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Alexis Diaz-Vegas
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Ariel Contreras-Ferrat
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Francisco Westermeier
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
| | - Enrique Jaimovich
- Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Chile
| | - Saverio Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas & Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Center for Studies of Exercise, Metabolism and Cancer (CEMC), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
38
|
Shahnoor N, Siebers EM, Brown KJ, Lawlor MW. Pathological Issues in Dystrophinopathy in the Age of Genetic Therapies. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:105-126. [PMID: 30148687 DOI: 10.1146/annurev-pathmechdis-012418-012945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dystrophinopathy is a class of genetic skeletal muscle disease characterized by myofiber degeneration and regeneration due to insufficient levels or functioning of dystrophin. Pathological evaluation for dystrophinopathy includes the identification of dystrophic skeletal muscle pathology and the immunohistochemical evaluation of dystrophin epitopes, but biopsies have become rare in recent years. However, the evaluation of dystrophin expression in the research setting has become critically important due to recent advances in genetic therapies, including exon skipping and gene therapy. Given the number of these therapies under evaluation in patients, it is likely that the traditional methods of evaluating dystrophinopathy will need to evolve in the near future. This review discusses current muscle biopsy diagnostic practices in dystrophinopathy and further focuses on how these practices have evolved in the context of therapeutic interventions for dystrophinopathy.
Collapse
Affiliation(s)
- Nazima Shahnoor
- Department of Pathology and Laboratory Medicine, and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA; , ,
| | - Emily M Siebers
- Department of Pathology and Laboratory Medicine, and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA; , ,
| | - Kristy J Brown
- Solid Biosciences, Inc., Cambridge, Massachusetts 02139, USA;
| | - Michael W Lawlor
- Department of Pathology and Laboratory Medicine, and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, USA; , ,
| |
Collapse
|
39
|
Guadagnin E, Mázala D, Chen YW. STAT3 in Skeletal Muscle Function and Disorders. Int J Mol Sci 2018; 19:ijms19082265. [PMID: 30072615 PMCID: PMC6121875 DOI: 10.3390/ijms19082265] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 07/19/2018] [Accepted: 07/23/2018] [Indexed: 12/17/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) signaling plays critical roles in regulating skeletal muscle mass, repair, and diseases. In this review, we discuss the upstream activators of STAT3 in skeletal muscles, with a focus on interleukin 6 (IL6) and transforming growth factor beta 1 (TGF-β1). We will also discuss the double-edged effect of STAT3 activation in the muscles, including the role of STAT3 signaling in muscle hypertrophy induced by exercise training or muscle wasting in cachectic diseases and muscular dystrophies. STAT3 is a critical regulator of satellite cell self-renewal after muscle injury. STAT3 knock out affects satellite cell myogenic progression by impairing proliferation and inducing premature differentiation. Recent studies in STAT3 signaling demonstrated its direct role in controlling myogenic capacity of myoblasts and satellite cells, as well as the potential benefit in using STAT3 inhibitors to treat muscle diseases. However, prolonged STAT3 activation in muscles has been shown to be responsible for muscle wasting by activating protein degradation pathways. It is important to balance the extent of STAT3 activation and the duration and location (cell types) of the STAT3 signaling when developing therapeutic interventions. STAT3 signaling in other tissues and organs that can directly or indirectly affects skeletal muscle health are also discussed.
Collapse
Affiliation(s)
- Eleonora Guadagnin
- Department of Orthopeadic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Davi Mázala
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA.
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA.
- Department Genomics and Precision Medicine, George Washington University, Washington, DC 20052, USA.
| |
Collapse
|
40
|
Nutrition in Duchenne muscular dystrophy 16–18 March 2018, Zaandam, the Netherlands. Neuromuscul Disord 2018; 28:680-689. [DOI: 10.1016/j.nmd.2018.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/09/2018] [Indexed: 11/17/2022]
|
41
|
Vellecco V, Armogida C, Bucci M. Hydrogen sulfide pathway and skeletal muscle: an introductory review. Br J Pharmacol 2018; 175:3090-3099. [PMID: 29767441 PMCID: PMC6031874 DOI: 10.1111/bph.14358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/18/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
The presence of the H2 S pathway in skeletal muscle (SKM) has recently been established. SKM expresses the three constitutive H2 S-generating enzymes in animals and humans, and it actively produces H2 S. The main, recognized molecular targets of H2 S, that is, potassium channels and PDEs, have been evaluated in SKM physiology in order to hypothesize a role for H2 S signalling. SKM dysfunctions, including muscular dystrophy and malignant hyperthermia, have also been evaluated as conditions in which the H2 S and transsulfuration pathways have been suggested to be involved. The intrinsic complexity of the molecular mechanisms involved in excitation-contraction (E-C) coupling together with the scarcity of preclinical models of SKM-related disorders have hampered any advances in the knowledge of SKM function. Here, we have addressed the role of the H2 S pathway in E-C coupling and the relative importance of cystathionine β-synthase, cistathionine γ-lyase and 3-mercaptopyruvate sulfurtransferase in SKM diseases.
Collapse
Affiliation(s)
- Valentina Vellecco
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Naples, 80131, Italy
| | - Chiara Armogida
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Naples, 80131, Italy
| | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine, University of Naples 'Federico II', Naples, 80131, Italy
| |
Collapse
|
42
|
Bulaklak K, Xiao B, Qiao C, Li J, Patel T, Jin Q, Li J, Xiao X. MicroRNA-206 Downregulation Improves Therapeutic Gene Expression and Motor Function in mdx Mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:283-293. [PMID: 30195767 PMCID: PMC6011022 DOI: 10.1016/j.omtn.2018.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a severe muscle-wasting disorder caused by a mutation in the dystrophin gene. Numerous gene therapies have been developed to replace or repair the defective dystrophin gene; however, these treatments cannot restore the full-length protein or completely resolve dystrophic symptoms. Secondary pathological mechanisms, such as functional ischemia and fibrosis, are thought to exacerbate the primary defect and cause the profound muscle degeneration found in dystrophic muscle. Surrogate therapies utilizing alternative therapeutic genes, or “booster genes,” such as VEGFA and utrophin, seek to address these secondary mechanisms and have shown impressive benefit in mdx mice. A skeletal muscle-specific microRNA, miR-206, is particularly overexpressed in dystrophic muscle and inhibits the expression of known booster genes. Thus, we aimed to determine if miR-206 contributes to dystrophic pathology by repressing beneficial gene expression. Here, we show that AAV-mediated expression of a miR-206 decoy target effectively downregulated miR-206 expression and increased endogenous therapeutic gene expression in mature mdx muscle. Furthermore, treatment significantly improved motor function and dystrophic pathology in mdx mice. In summary, we have identified a contributing factor to the dystrophic phenotype and characterized a novel therapeutic avenue for DMD.
Collapse
Affiliation(s)
- Karen Bulaklak
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bin Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Chunping Qiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jianbin Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tejash Patel
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Quan Jin
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juan Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xiao Xiao
- Division of Pharmacoengineering and Molecular Pharmaceutics, Department of Pharmaceutical Sciences, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
43
|
van Putten M, Aartsma-Rus A, Grounds MD, Kornegay JN, Mayhew A, Gillingwater TH, Takeda S, Rüegg MA, De Luca A, Nagaraju K, Willmann R. Update on Standard Operating Procedures in Preclinical Research for DMD and SMA Report of TREAT-NMD Alliance Workshop, Schiphol Airport, 26 April 2015, The Netherlands. J Neuromuscul Dis 2018; 5:29-34. [PMID: 29480217 PMCID: PMC5836406 DOI: 10.3233/jnd-170288] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A workshop took place in 2015 to follow up TREAT-NMD activities dedicated to improving quality in the preclinical phase of drug development for neuromuscular diseases. In particular, this workshop adressed necessary future steps regarding common standard experimental protocols and the issue of improving the translatability of preclinical efficacy studies.
Collapse
Affiliation(s)
- Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Miranda D Grounds
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, WA, Australia
| | - Joe N Kornegay
- College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, TX, USA
| | - Anna Mayhew
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Shin'ichi Takeda
- National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | | | - Annamaria De Luca
- Department of Pharmacy and Drug Sciences, Unit of Pharmacology, University of Bari, Bari, Italy
| | | | - Raffaella Willmann
- Biozentrum, University of Basel, Basel, Switzerland.,Swiss Foundation for Research on Muscle Diseases, Cortaillod, Switzerland
| |
Collapse
|
44
|
Perkins KJ, Davies KE. Alternative utrophin mRNAs contribute to phenotypic differences between dystrophin-deficient mice and Duchenne muscular dystrophy. FEBS Lett 2018; 592:1856-1869. [PMID: 29772070 PMCID: PMC6032923 DOI: 10.1002/1873-3468.13099] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/01/2018] [Accepted: 05/07/2018] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal disorder caused by absence of functional dystrophin protein. Compensation in dystrophin‐deficient (mdx) mice may be achieved by overexpression of its fetal paralogue, utrophin. Strategies to increase utrophin levels by stimulating promoter activity using small compounds are therefore a promising pharmacological approach. Here, we characterise similarities and differences existing within the mouse and human utrophin locus to assist in high‐throughput screening for potential utrophin modulator drugs. We identified five novel 5′‐utrophin isoforms (A′,B′,C,D and F) in adult and embryonic tissue. As the more efficient utrophin‐based response in mdx skeletal muscle appears to involve independent transcriptional activation of conserved, myogenic isoforms (A′ and F), elevating their paralogues in DMD patients is an encouraging therapeutic strategy.
Collapse
Affiliation(s)
- Kelly J Perkins
- Department of Physiology Anatomy and Genetics, University of Oxford, UK.,Sir William Dunn School of Pathology, University of Oxford, UK
| | - Kay E Davies
- Department of Physiology Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
45
|
Charles JP, Cappellari O, Hutchinson JR. A Dynamic Simulation of Musculoskeletal Function in the Mouse Hindlimb During Trotting Locomotion. Front Bioeng Biotechnol 2018; 6:61. [PMID: 29868576 PMCID: PMC5964171 DOI: 10.3389/fbioe.2018.00061] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 11/30/2022] Open
Abstract
Mice are often used as animal models of various human neuromuscular diseases, and analysis of these models often requires detailed gait analysis. However, little is known of the dynamics of the mouse musculoskeletal system during locomotion. In this study, we used computer optimization procedures to create a simulation of trotting in a mouse, using a previously developed mouse hindlimb musculoskeletal model in conjunction with new experimental data, allowing muscle forces, activation patterns, and levels of mechanical work to be estimated. Analyzing musculotendon unit (MTU) mechanical work throughout the stride allowed a deeper understanding of their respective functions, with the rectus femoris MTU dominating the generation of positive and negative mechanical work during the swing and stance phases. This analysis also tested previous functional inferences of the mouse hindlimb made from anatomical data alone, such as the existence of a proximo-distal gradient of muscle function, thought to reflect adaptations for energy-efficient locomotion. The results do not strongly support the presence of this gradient within the mouse musculoskeletal system, particularly given relatively high negative net work output from the ankle plantarflexor MTUs, although more detailed simulations could test this further. This modeling analysis lays a foundation for future studies of the control of vertebrate movement through the development of neuromechanical simulations.
Collapse
Affiliation(s)
- James P Charles
- Neuromuscular Diseases Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,Structure and Motion Lab, Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, United Kingdom
| | - Ornella Cappellari
- Neuromuscular Diseases Group, Department of Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom
| | - John R Hutchinson
- Structure and Motion Lab, Department of Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, United Kingdom
| |
Collapse
|
46
|
Kalra S, Montanaro F, Denning C. Can Human Pluripotent Stem Cell-Derived Cardiomyocytes Advance Understanding of Muscular Dystrophies? J Neuromuscul Dis 2018; 3:309-332. [PMID: 27854224 PMCID: PMC5123622 DOI: 10.3233/jnd-150133] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Muscular dystrophies (MDs) are clinically and molecularly a highly heterogeneous group of single-gene disorders that primarily affect striated muscles. Cardiac disease is present in several MDs where it is an important contributor to morbidity and mortality. Careful monitoring of cardiac issues is necessary but current management of cardiac involvement does not effectively protect from disease progression and cardiac failure. There is a critical need to gain new knowledge on the diverse molecular underpinnings of cardiac disease in MDs in order to guide cardiac treatment development and assist in reaching a clearer consensus on cardiac disease management in the clinic. Animal models are available for the majority of MDs and have been invaluable tools in probing disease mechanisms and in pre-clinical screens. However, there are recognized genetic, physiological, and structural differences between human and animal hearts that impact disease progression, manifestation, and response to pharmacological interventions. Therefore, there is a need to develop parallel human systems to model cardiac disease in MDs. This review discusses the current status of cardiomyocytes (CMs) derived from human induced pluripotent stem cells (hiPSC) to model cardiac disease, with a focus on Duchenne muscular dystrophy (DMD) and myotonic dystrophy (DM1). We seek to provide a balanced view of opportunities and limitations offered by this system in elucidating disease mechanisms pertinent to human cardiac physiology and as a platform for treatment development or refinement.
Collapse
Affiliation(s)
- Spandan Kalra
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| | - Federica Montanaro
- Dubowitz Neuromuscular Centre, Department of Molecular Neurosciences, University College London - Institute of Child Health, London, UK
| | - Chris Denning
- Department of Stem Cell Biology, Centre for Biomolecular Sciences, University of Nottingham, UK
| |
Collapse
|
47
|
van der Bent ML, Paulino da Silva Filho O, van Luijk J, Brock R, Wansink DG. Assisted delivery of antisense therapeutics in animal models of heritable neurodegenerative and neuromuscular disorders: a systematic review and meta-analysis. Sci Rep 2018; 8:4181. [PMID: 29520012 PMCID: PMC5843643 DOI: 10.1038/s41598-018-22316-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Antisense oligonucleotide (AON)-based therapies hold promise for a range of neurodegenerative and neuromuscular diseases and have shown benefit in animal models and patients. Success in the clinic is nevertheless still limited, due to unfavourable biodistribution and poor cellular uptake of AONs. Extensive research is currently being conducted into the formulation of AONs to improve delivery, but thus far there is no consensus on which of those strategies will be the most effective. This systematic review was designed to answer in an unbiased manner which delivery strategies most strongly enhance the efficacy of AONs in animal models of heritable neurodegenerative and neuromuscular diseases. In total, 95 primary studies met the predefined inclusion criteria. Study characteristics and data on biodistribution and toxicity were extracted and reporting quality and risk of bias were assessed. Twenty studies were eligible for meta-analysis. We found that even though the use of delivery systems provides an advantage over naked AONs, it is not yet possible to select the most promising strategies. Importantly, standardisation of experimental procedures is warranted in order to reach conclusions about the most efficient delivery strategies. Our best practice guidelines for future experiments serve as a step in that direction.
Collapse
Affiliation(s)
- M Leontien van der Bent
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - Omar Paulino da Silva Filho
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
- CAPES Foundation, Ministry of Education of Brazil, Brasília, Brazil
| | - Judith van Luijk
- Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE), Department of Health Evidence, Radboud university medical center, Nijmegen, The Netherlands
| | - Roland Brock
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands
| | - Derick G Wansink
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud university medical center, Nijmegen, The Netherlands.
| |
Collapse
|
48
|
Capogrosso RF, Mantuano P, Uaesoontrachoon K, Cozzoli A, Giustino A, Dow T, Srinivassane S, Filipovic M, Bell C, Vandermeulen J, Massari AM, De Bellis M, Conte E, Pierno S, Camerino GM, Liantonio A, Nagaraju K, De Luca A. Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy. FASEB J 2018; 32:1025-1043. [PMID: 29097503 PMCID: PMC5888399 DOI: 10.1096/fj.201700182rrr] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 10/16/2017] [Indexed: 12/19/2022]
Abstract
Muscle fibers lacking dystrophin undergo a long-term alteration of Ca2+ homeostasis, partially caused by a leaky Ca2+ release ryanodine (RyR) channel. S48168/ARM210, an RyR calcium release channel stabilizer (a Rycal compound), is expected to enhance the rebinding of calstabin to the RyR channel complex and possibly alleviate the pathologic Ca2+ leakage in dystrophin-deficient skeletal and cardiac muscle. This study systematically investigated the effect of S48168/ARM210 on the phenotype of mdx mice by means of a first proof-of-concept, short (4 wk), phase 1 treatment, followed by a 12-wk treatment (phase 2) performed in parallel by 2 independent laboratories. The mdx mice were treated with S48168/ARM210 at two different concentrations (50 or 10 mg/kg/d) in their drinking water for 4 and 12 wk, respectively. The mice were subjected to treadmill sessions twice per week (12 m/min for 30 min) to unmask the mild disease. This testing was followed by in vivo forelimb and hindlimb grip strength and fatigability measurement, ex vivo extensor digitorum longus (EDL) and diaphragm (DIA) force contraction measurement and histologic and biochemical analysis. The treatments resulted in functional (grip strength, ex vivo force production in DIA and EDL muscles) as well as histologic improvement after 4 and 12 wk, with no adverse effects. Furthermore, levels of cellular biomarkers of calcium homeostasis increased. Therefore, these data suggest that S48168/ARM210 may be a safe therapeutic option, at the dose levels tested, for the treatment of Duchenne muscular dystrophy (DMD).-Capogrosso, R. F., Mantuano, P., Uaesoontrachoon, K., Cozzoli, A., Giustino, A., Dow, T., Srinivassane, S., Filipovic, M., Bell, C., Vandermeulen, J., Massari, A. M., De Bellis, M., Conte, E., Pierno, S., Camerino, G. M., Liantonio, A., Nagaraju, K., De Luca, A. Ryanodine channel complex stabilizer compound S48168/ARM210 as a disease modifier in dystrophin-deficient mdx mice: proof-of-concept study and independent validation of efficacy.
Collapse
Affiliation(s)
| | - Paola Mantuano
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | | | - Anna Cozzoli
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Arcangela Giustino
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Todd Dow
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | | | - Marina Filipovic
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | - Christina Bell
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
| | | | - Ada Maria Massari
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Michela De Bellis
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Elena Conte
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Sabata Pierno
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Giulia Maria Camerino
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Antonella Liantonio
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| | - Kanneboyina Nagaraju
- Agada Biosciences Incorporated, Halifax, Nova Scotia, Canada; and
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, Binghamton, New York, USA
| | - Annamaria De Luca
- Pharmacology Unit, Department of Pharmacy–Drug Sciences, University of Bari, Bari, Italy
| |
Collapse
|
49
|
Abnormal carbohydrate metabolism in a canine model for muscular dystrophy. J Nutr Sci 2017; 6:e57. [PMID: 29209496 PMCID: PMC5705810 DOI: 10.1017/jns.2017.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/26/2017] [Accepted: 09/28/2017] [Indexed: 11/21/2022] Open
Abstract
The canine golden retriever muscular dystrophy (GRMD) model is the best animal model for studying Duchenne muscular dystrophy in humans. Considering the importance of glucose metabolism in the muscles, the existence of metabolic and endocrine alterations in a wide range of muscular dystrophies, and the pre-existing relationship between blood insulin concentration and muscular atrophy, the present study aimed to evaluate the postprandial glucose and insulin response in GRMD dogs. A total of eighteen golden retriever dogs were randomly distributed into three experimental groups: healthy/control (G1), female GRMD carriers (G2), and male dogs affected by GRMD (G3). Higher plasma resting glucose levels (P = 0·0047) were seen in G2 and G3 compared with G1, as was the case for minimum (P = <0·0001), mean (P = 0·0002) and maximum (P = 0·0359) glucose values for G3 compared with G1. Fructosamine concentrations were in accordance with reference values found in the literature for dogs. Insulin levels were lower in G3 compared with G1 (P = 0·0065); however, there was no evidence of insulin resistance according to the homeostasis model assessment index values obtained. As for the evaluation of postprandial responses, fluctuations of glucose (P = 0·0007) and insulin (P = 0·0149) were observed in G1 and G2, while in G3 the values remained constant. The results allowed us to identify metabolic changes related to carbohydrate metabolism in GRMD dogs, highlighting the importance of adequate food management for these animals.
Collapse
|
50
|
Hu X, Charles JP, Akay T, Hutchinson JR, Blemker SS. Are mice good models for human neuromuscular disease? Comparing muscle excursions in walking between mice and humans. Skelet Muscle 2017; 7:26. [PMID: 29145886 PMCID: PMC5689180 DOI: 10.1186/s13395-017-0143-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 10/26/2017] [Indexed: 11/29/2022] Open
Abstract
Background The mouse is one of the most widely used animal models to study neuromuscular diseases and test new therapeutic strategies. However, findings from successful pre-clinical studies using mouse models frequently fail to translate to humans due to various factors. Differences in muscle function between the two species could be crucial but often have been overlooked. The purpose of this study was to evaluate and compare muscle excursions in walking between mice and humans. Methods Recently published musculoskeletal models of the mouse hindlimb and human lower limb were used to simulate muscle-tendon dynamics during mouse and human walking, a key daily activity. Muscle fiber length changes (fiber excursions) of 25 muscle homologs in the two species were calculated from these simulations and then compared. To understand potential causes of differences in fiber excursions in walking, joint excursions and muscle moment arms were also compared across one gait cycle. Results Most muscles (19 out of 25 muscles) of the mouse hindlimb had much smaller fiber excursions as compared to human lower limb muscles during walking. For these muscles, fiber excursions in mice were only 48 ± 19% of those in humans. The differences in fiber excursion between the two species were primarily due to the reduced joint excursions and smaller muscle moment arms in mice as compared to humans. Conclusions Since progressive neuromuscular diseases, such as Duchenne muscular dystrophy, are known to be accelerated by damage accumulated from active muscle lengthening, these results suggest that biomechanical differences in muscle function during walking between mice and humans may impede the translations of knowledge gained from mouse models to humans. This knowledge would add a fresh perspective on how pre-clinical studies on mice might be better designed to improve translation to human clinical trials. Electronic supplementary material The online version of this article (10.1186/s13395-017-0143-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao Hu
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, 22908, USA
| | - James P Charles
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Turgay Akay
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - John R Hutchinson
- Comparative Biomedical Sciences, Royal Veterinary College, Hatfield, Hertfordshire, AL9 7TA, UK
| | - Silvia S Blemker
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road, Charlottesville, VA, 22908, USA. .,Department of Orthopaedic Surgery, University of Virginia, Charlottesville, VA, 22903, USA. .,Department of Mechanical and Aerospace Engineering, University of Virginia, Charlottesville, VA, 22903, USA.
| |
Collapse
|