51
|
Ponzetti M, Aielli F, Ucci A, Cappariello A, Lombardi G, Teti A, Rucci N. Lipocalin 2 increases after high-intensity exercise in humans and influences muscle gene expression and differentiation in mice. J Cell Physiol 2021; 237:551-565. [PMID: 34224151 PMCID: PMC9291458 DOI: 10.1002/jcp.30501] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 06/04/2021] [Accepted: 06/25/2021] [Indexed: 12/22/2022]
Abstract
Lipocalin 2 (LCN2) is an adipokine that accomplishes several functions in diverse organs. However, its importance in muscle and physical exercise is currently unknown. We observed that following acute high‐intensity exercise (“Gran Sasso d'Italia” vertical run), LCN2 serum levels were increased. The Wnt pathway antagonist, DKK1, was also increased after the run, positively correlating with LCN2, and the same was found for the cytokine Interleukin 6. We, therefore, investigated the involvement of LCN2 in muscle physiology employing an Lcn2 global knockout (Lcn2−/−) mouse model. Lcn2−/− mice presented with smaller muscle fibres but normal muscle performance (grip strength metre) and muscle weight. At variance with wild type (WT) mice, the inflammatory cytokine Interleukin 6 was undetectable in Lcn2−/− mice at all ages. Intriguingly, Lcn2−/− mice did not lose gastrocnemius and quadriceps muscle mass and muscle performance following hindlimb suspension, while at variance with WT, they lose soleus muscle mass. In vitro, LCN2 treatment reduced the myogenic differentiation of C2C12 and primary mouse myoblasts and influenced their gene expression. Treating myoblasts with LCN2 reduced myogenesis, suggesting that LCN2 may negatively affect muscle physiology when upregulated following high‐intensity exercise.
Collapse
Affiliation(s)
- Marco Ponzetti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Federica Aielli
- Medical Oncology Department, Giuseppe Mazzini Hospital, Teramo, Italy
| | - Argia Ucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Alfredo Cappariello
- Research Laboratories, Department of Onco-haematology, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy.,Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Nadia Rucci
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
52
|
Ban J, Beqaj B, Phillips WD. Vector-mediated expression of muscle specific kinase restores specific force to muscles in the mdx mouse model of Duchenne muscular dystrophy. Exp Physiol 2021; 106:1794-1805. [PMID: 34114278 DOI: 10.1113/ep089439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/08/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? The (dystrophin-deficient) muscles of mdx mice generate less contractile force per cross-sectional area (specific force) than those of healthy wild-type mice: what is the influence of muscle specific kinase (MuSK) upon the properties of the tibialis anterior (TA) muscle in mdx mice? What is the main finding and its importance? Injection of adeno-associated viral vector encoding MuSK into the TA muscle of young mdx mice increased the specific force of the muscle, suggesting the MuSK signalling system has the potential to restore healthy growth to dystrophin-deficient muscles. ABSTRACT In the mdx mouse model of Duchenne muscular dystrophy, muscle fibres are fragile and prone to injury and degeneration. Compared to wild-type mice, muscles of mdx mice also develop less specific force (contractile force/cross-sectional area). We recently reported that injecting adeno-associated viral vector encoding muscle specific kinase (AAV-MuSK) into muscles of mdx mice increased utrophin expression and made the muscles more resistant to acute stretch-induced injury. Here we injected AAV-MuSK unilaterally into the tibialis anterior muscle of mdx mice at a younger age (4 weeks), and recorded contraction force from the muscles in situ at 12 weeks of age. Compared to contralateral empty-vector control muscles, muscles injected with AAV-MuSK produced 28% greater specific force (P = 0.0005). They did not undergo the compensatory hypertrophy that normally occurs in muscles of mdx mice. Injection of AAV encoding rapsyn (a downstream effector of MuSK signalling) caused no such improvement in muscle strength. Muscles injected with AAV-MuSK displayed a 10% reduction in the number of fibres with centralized nuclei (P = 0.0015). Our results in mdx mice suggest that elevating the expression of MuSK can reduce the incidence of muscle fibre regeneration and improve the strength of dystrophin-deficient muscles.
Collapse
Affiliation(s)
- Joanne Ban
- Physiology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Besa Beqaj
- Physiology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - William D Phillips
- Physiology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
53
|
Mitchell R, Frederick NE, Holzman ER, Agobe F, Allaway HCM, Bagher P. Ifetroban reduces coronary artery dysfunction in a mouse model of Duchenne muscular dystrophy. Am J Physiol Heart Circ Physiol 2021; 321:H52-H58. [PMID: 34048282 DOI: 10.1152/ajpheart.00180.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Dilated cardiomyopathy contributes to morbidity and mortality in Duchenne muscular dystrophy (DMD), an inheritable muscle-wasting disease caused by a mutation in the dystrophin gene. Preclinical studies in mouse models of muscular dystrophy have demonstrated reduced cardiomyopathy and improved cardiac function following oral treatment with the potent and selective thromboxane A2/prostanoid receptor (TPr) antagonist ifetroban. Furthermore, a phase 2 clinical trial (NCT03340675, Cumberland Pharmaceuticals) is currently recruiting subjects to determine whether ifetroban can improve cardiac function in patients with DMD. Although TPr is a promising therapeutic target for the treatment of dilated cardiomyopathy in DMD, little is known about TPr function in coronary arteries that perfuse blood through the cardiac tissue. In the current study, isolated coronary arteries from young (∼3-5 mo) and aged (∼9-12 mo) mdx mice, a widely used mouse model of DMD, and age-matched controls were examined using wire myography. Vasoconstriction to increasing concentrations of TPr agonist U-46619 (U4) was enhanced in young mdx mice versus controls. In addition, young mdx mice displayed a significant attenuation in endothelial cell-mediated vasodilation to increasing concentrations of the muscarinic agonist acetylcholine (ACh). Since TPr activation was enhanced in young mdx mice, U4-mediated vasoconstriction was measured in the absence and the presence of ifetroban. Ifetroban reduced U4-mediated vasoconstriction in young mdx mice and both aged mdx and control mice. Overall, our data demonstrate enhanced coronary arterial vasoconstriction to TPr activation in young mdx mice, a phenotype that could be reversed with ifetroban. These data could have important therapeutic implications for improving cardiovascular function in DMD.NEW & NOTEWORTHY This investigation revealed 1) impaired acetylcholine-mediated vasodilation, 2) increased U-46619-mediated vasoconstriction, and 3) reversal of the increase in U-46619-mediated vasoconstriction by the thromboxane A2/prostanoid receptor (TPr) antagonist ifetroban in left anterior descending coronary arteries isolated from young mdx mice, a model of Duchenne muscular dystrophy (DMD). Ifetroban has been used in preclinical studies to demonstrate improved cardiac function in mouse models of muscular dystrophy and is currently being investigated in a phase 2 clinical trial in patients with DMD. The current study supports the role of ifetroban in improving coronary artery function in preclinical DMD models, which may contribute to improved cardiovascular health.
Collapse
Affiliation(s)
- R Mitchell
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - N E Frederick
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - E R Holzman
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - F Agobe
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - H C M Allaway
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - P Bagher
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, Texas
| |
Collapse
|
54
|
Dudley RWR, Comtois AS, St-Pierre DH, Danialou G. Early administration of L-arginine in mdx neonatal mice delays the onset of muscular dystrophy in tibialis anterior (TA) muscle. FASEB Bioadv 2021; 3:639-651. [PMID: 34377959 PMCID: PMC8332474 DOI: 10.1096/fba.2020-00104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 02/17/2021] [Accepted: 04/15/2021] [Indexed: 12/04/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder that results in the absence of dystrophin, a cytoskeletal protein. Individuals with this disease experience progressive muscle destruction, which leads to muscle weakness. Studies have been conducted to find solutions for the relief of individuals with this disease, several of which have shown that utrophin, a protein closely related to dystrophin, when overexpressed in mdx neonatal mice (the murine model of DMD), is able to prevent the progressive muscle destruction observed in the absence of dystrophin. Furthermore, recent studies have shown that L‐arginine induces utrophin upregulation in adult mdx mice. We hypothesized that L‐arginine treatment also induces utrophin upregulation to prevent the development of muscle weakness in neonatal mdx mice. Hence, L‐arginine should also prevent progressive muscle destruction via utrophin upregulation in mdx neonatal mice. Mdx neonatal mice were injected intraperitoneally daily with 800 mg/kg of L‐arginine for 6 weeks, whereas control mice were injected with a physiological saline. The following experiments were performed on the tibialis anterior (TA) muscle: muscle contractility and resistance to mechanical stress; central nucleation and peripheral nucleation, utrophin, and creatine kinase quantification as well as a nitric oxide (NO) assay. Our findings show that early administration of L‐arginine in mdx neonatal mice prevents the destruction of the tibialis anterior (TA) muscle. However, this improvement was related to nitric oxide (NO) production rather than the expected utrophin upregulation.
Collapse
Affiliation(s)
- Roy W R Dudley
- Meakins Christie Laboratories McGill University Montreal QC Canada
| | - Alain S Comtois
- Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Groupe de Recherche en Activité Physique Adaptée UQAM Montreal QC Canada
| | - David H St-Pierre
- Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Groupe de Recherche en Activité Physique Adaptée UQAM Montreal QC Canada.,Centre de Recherche du CHU Sainte-Justine Montréal QC Canada
| | - Gawiyou Danialou
- Meakins Christie Laboratories McGill University Montreal QC Canada.,Département des Sciences de l'Activité Physique Université du Québec à Montréal (UQAM Montreal QC Canada.,Royal Military College Saint-Jean Saint-Jean-sur-Richelieu QC Canada
| |
Collapse
|
55
|
Soblechero-Martín P, López-Martínez A, de la Puente-Ovejero L, Vallejo-Illarramendi A, Arechavala-Gomeza V. Utrophin modulator drugs as potential therapies for Duchenne and Becker muscular dystrophies. Neuropathol Appl Neurobiol 2021; 47:711-723. [PMID: 33999469 PMCID: PMC8518368 DOI: 10.1111/nan.12735] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/28/2021] [Accepted: 05/10/2021] [Indexed: 12/25/2022]
Abstract
Utrophin is an autosomal paralogue of dystrophin, a protein whose deficit causes Duchenne and Becker muscular dystrophies (DMD/BMD). Utrophin is naturally overexpressed at the sarcolemma of mature dystrophin‐deficient fibres in DMD and BMD patients as well as in the mdx Duchenne mouse model. Dystrophin and utrophin can co‐localise in human foetal muscle, in the dystrophin‐competent fibres from DMD/BMD carriers, and revertant fibre clusters in biopsies from DMD patients. These findings suggest that utrophin overexpression could act as a surrogate, compensating for the lack of dystrophin, and, as such, it could be used in combination with dystrophin restoration therapies. Different strategies to overexpress utrophin are currently under investigation. In recent years, many compounds have been reported to modulate utrophin expression efficiently in preclinical studies and ameliorate the dystrophic phenotype in animal models of the disease. In this manuscript, we discuss the current knowledge on utrophin protein and the different mechanisms that modulate its expression in skeletal muscle. We also include a comprehensive review of compounds proposed as utrophin regulators and, as such, potential therapeutic candidates for these muscular dystrophies.
Collapse
Affiliation(s)
- Patricia Soblechero-Martín
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Clinical Laboratory Service, Osakidetza Basque Health Service, Bilbao-Basurto Integrated Health Organisation, Basurto University Hospital, Bilbao, Spain
| | - Andrea López-Martínez
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | | | | | - Virginia Arechavala-Gomeza
- Neuromuscular Disorders, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
56
|
Uryash A, Mijares A, Esteve E, Adams JA, Lopez JR. Cardioprotective Effect of Whole Body Periodic Acceleration in Dystrophic Phenotype mdx Rodent. Front Physiol 2021; 12:658042. [PMID: 34017265 PMCID: PMC8129504 DOI: 10.3389/fphys.2021.658042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/09/2021] [Indexed: 01/14/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle wasting and the development of a dilated cardiomyopathy (DCM), which is the leading cause of death in DMD patients. Despite knowing the cause of DMD, there are currently no therapies which can prevent or reverse its inevitable progression. We have used whole body periodic acceleration (WBPA) as a novel tool to enhance intracellular constitutive nitric oxide (NO) production. WBPA adds small pulses to the circulation to increase pulsatile shear stress, thereby upregulating endothelial nitric oxide synthase (eNOS) and neuronal nitric oxide synthase (nNOS) and subsequently elevating the production of NO. Myocardial cells from dystrophin-deficient 15-month old mdx mice have contractile deficiency, which is associated with elevated concentrations of diastolic Ca2+ ([Ca2+]d), Na+ ([Na+]d), and reactive oxygen species (ROS), increased cell injury, and decreased cell viability. Treating 12-month old mdx mice with WBPA for 3 months reduced cardiomyocyte [Ca2+]d and [Na+]d overload, decreased ROS production, and upregulated expression of the protein utrophin resulting in increased cell viability, reduced cardiomyocyte damage, and improved contractile function compared to untreated mdx mice.
Collapse
Affiliation(s)
- Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Alfredo Mijares
- Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas, Caracas, Venezuela
| | - Eric Esteve
- UMR 5525 UGA-CNRS-Grenoble INP-VetAgro Sup TIMC, Université Grenoble Alpes, Grenoble, France
| | - Jose A Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL, United States
| | - Jose R Lopez
- Department of Molecular Biosciences, University of California, Davis, Davis, CA, United States.,Department of Research, Mount Sinai Medical Center, Miami Beach, FL, United States
| |
Collapse
|
57
|
Howard ZM, Dorn LE, Lowe J, Gertzen MD, Ciccone P, Rastogi N, Odom GL, Accornero F, Chamberlain JS, Rafael-Fortney JA. Micro-dystrophin gene therapy prevents heart failure in an improved Duchenne muscular dystrophy cardiomyopathy mouse model. JCI Insight 2021; 6:146511. [PMID: 33651713 PMCID: PMC8119181 DOI: 10.1172/jci.insight.146511] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/24/2021] [Indexed: 12/19/2022] Open
Abstract
Gene replacement for Duchenne muscular dystrophy (DMD) with micro-dystrophins has entered clinical trials, but efficacy in preventing heart failure is unknown. Although most patients with DMD die from heart failure, cardiomyopathy is undetectable until the teens, so efficacy from trials in young boys will be unknown for a decade. Available DMD animal models were sufficient to demonstrate micro-dystrophin efficacy on earlier onset skeletal muscle pathology underlying loss of ambulation and respiratory insufficiency in patients. However, no mouse models progressed into heart failure, and dog models showed highly variable progression insufficient to evaluate efficacy of micro-dystrophin or other therapies on DMD heart failure. To overcome this barrier, we have generated the first DMD mouse model to our knowledge that reproducibly progresses into heart failure. This model shows cardiac inflammation and fibrosis occur prior to reduced function. Fibrosis does not continue to accumulate, but inflammation persists after function declines. We used this model to test micro-dystrophin gene therapy efficacy on heart failure prevention for the first time. Micro-dystrophin prevented declines in cardiac function and prohibited onset of inflammation and fibrosis. This model will allow identification of committed pathogenic steps to heart failure and testing of genetic and nongenetic therapies to optimize cardiac care for patients with DMD.
Collapse
Affiliation(s)
- Zachary M. Howard
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Lisa E. Dorn
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jeovanna Lowe
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Megan D. Gertzen
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Pierce Ciccone
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Neha Rastogi
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Guy L. Odom
- Department of Neurology and Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, USA
| | - Federica Accornero
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jeffrey S. Chamberlain
- Department of Neurology and Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, USA
| | - Jill A. Rafael-Fortney
- Department of Physiology & Cell Biology and Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
58
|
Starosta A, Konieczny P. Therapeutic aspects of cell signaling and communication in Duchenne muscular dystrophy. Cell Mol Life Sci 2021; 78:4867-4891. [PMID: 33825942 PMCID: PMC8233280 DOI: 10.1007/s00018-021-03821-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/26/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating chromosome X-linked disease that manifests predominantly in progressive skeletal muscle wasting and dysfunctions in the heart and diaphragm. Approximately 1/5000 boys and 1/50,000,000 girls suffer from DMD, and to date, the disease is incurable and leads to premature death. This phenotypic severity is due to mutations in the DMD gene, which result in the absence of functional dystrophin protein. Initially, dystrophin was thought to be a force transducer; however, it is now considered an essential component of the dystrophin-associated protein complex (DAPC), viewed as a multicomponent mechanical scaffold and a signal transduction hub. Modulating signal pathway activation or gene expression through epigenetic modifications has emerged at the forefront of therapeutic approaches as either an adjunct or stand-alone strategy. In this review, we propose a broader perspective by considering DMD to be a disease that affects myofibers and muscle stem (satellite) cells, as well as a disorder in which abrogated communication between different cell types occurs. We believe that by taking this systemic view, we can achieve safe and holistic treatments that can restore correct signal transmission and gene expression in diseased DMD tissues.
Collapse
Affiliation(s)
- Alicja Starosta
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Patryk Konieczny
- Faculty of Biology, Institute of Human Biology and Evolution, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
59
|
Wu H, Hossain MM, Kim H, Gallippi CM, Jiang X. A 1.5-D Array for Acoustic Radiation Force (ARF)-Induced Peak Displacement-Based Tissue Anisotropy Assessment With a Row-Column Excitation Method. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:1278-1287. [PMID: 33044921 PMCID: PMC8080255 DOI: 10.1109/tuffc.2020.3030040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Many biological tissues, including muscle or kidney, are mechanically anisotropic, and the degree of anisotropy (DoA) in mechanical properties is diagnostically relevant. DoA can be assessed either using the ratio of shear wave velocities (SWVs) or acoustic radio forced impulse (ARFI)-induced peak displacements (PD) measured longitudinal over transverse orientations. Whether using SWV or PD as a basis, DoA expressed as the ratio of values requires 90° transducer rotation when a linear array is employed. This large rotation angle is prone to misalignment errors. One solution is the use of a fully sampled matrix array for electronic rotation of point spread function (PSF). However, the challenges of matrix array are its high fabrication cost and complicated fabrication procedures. The cheaper and simpler alternative of matrix array is the use of a row-column array. A 3×64 elements 1.5-D array with a row-column excitation mode is proposed to assess DoA in mechanical properties using the PD ratio. Different numbers of elements in elevational and lateral directions were selected to have orthogonal ARFI excitation beams without rotating the transducer. A custom-designed flex circuit was used to fabricate the array with a simpler electrode connection than a fully sampled matrix array. The performance of the array was evaluated in Field II simulation and experiment. The output pressure was 0.57-MPa output under a 40- [Formula: see text] excitation with a -6-dB point spread dimension of 14×4 mm2 in orthogonal directions. The PD was measured to be [Formula: see text] in an isotropic elastic phantom with Young's modulus of 5.4 kPa. These results suggest that the array is capable of assessing DoA using PD ratio without physical rotation of the transducer. The array has the potential to reduce the misalignment errors for DoA assessment.
Collapse
Affiliation(s)
- Huaiyu Wu
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Md Murad Hossain
- Department of Biomedical Engineering, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, NC 27599 USA, and North Carolina State University (NCSU), Raleigh, NC 27695 USA. He is now with the Department of Biomedical Engineering, Columbia University, New York, NY 10027 USA
| | - Howuk Kim
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695, USA
| | - Caterina M. Gallippi
- Joint Department of Biomedical Engineering, University of North Carolina (UNC) at Chapel Hill, Chapel Hill, NC 27599 USA, and North Carolina State University (NCSU), Raleigh, NC 27695 USA
| | - Xiaoning Jiang
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
60
|
Lindsay A, Kemp B, Larson AA, Baumann CW, McCourt PM, Holm J, Karachunski P, Lowe DA, Ervasti JM. Tetrahydrobiopterin synthesis and metabolism is impaired in dystrophin-deficient mdx mice and humans. Acta Physiol (Oxf) 2021; 231:e13627. [PMID: 33580591 DOI: 10.1111/apha.13627] [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: 11/29/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022]
Abstract
AIM Loss of dystrophin causes oxidative stress and affects nitric oxide synthase-mediated vascular function in striated muscle. Because tetrahydrobiopterin is an antioxidant and co-factor for nitric oxide synthase, we tested the hypothesis that tetrahydrobiopterin would be low in mdx mice and humans deficient for dystrophin. METHODS Tetrahydrobiopterin and its metabolites were measured at rest and in response to exercise in Duchenne and Becker muscular dystrophy patients, age-matched male controls as well as wild-type, mdx and mdx mice transgenically overexpressing skeletal muscle-specific dystrophins. Mdx mice were also supplemented with tetrahydrobiopterin and pathophysiology was assessed. RESULTS Duchenne muscular dystrophy patients had lower urinary dihydrobiopterin + tetrahydrobiopterin/specific gravity1.020 compared to unaffected age-matched males and Becker muscular dystrophy patients. Mdx mice had low urinary and skeletal muscle dihydrobiopterin + tetrahydrobiopterin compared to wild-type mice. Overexpression of dystrophins that localize neuronal nitric oxide synthase restored dihydrobiopterin + tetrahydrobiopterin in mdx mice to wild-type levels while utrophin overexpression did not. Mdx mice and Duchenne muscular dystrophy patients did not increase tetrahydrobiopterin during exercise and in mdx mice tetrahydrobiopterin deficiency was likely because of lower levels of sepiapterin reductase in skeletal muscle. Tetrahydrobiopterin supplementation improved skeletal muscle strength, resistance to fatiguing and injurious contractions in vivo, increased utrophin and capillary density of skeletal muscle and lowered cardiac muscle fibrosis and left ventricular wall thickness in mdx mice. CONCLUSION These data demonstrate that impaired tetrahydrobiopterin synthesis is associated with dystrophin loss and treatment with tetrahydrobiopterin improves striated muscle histopathology and skeletal muscle function in mdx mice.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Bailey Kemp
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexie A Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Cory W Baumann
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Preston M McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - John Holm
- Lillehei Heart Institute, Cancer and Cardiovascular Research Center, University of Minnesota, Minneapolis, MN, USA
| | - Peter Karachunski
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Department of Neurology, University of Minnesota, Minneapolis, MN, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
61
|
Martin PT, Zygmunt DA, Ashbrook A, Hamilton S, Packer D, Birch SM, Bettis AK, Balog-Alvarez CJ, Guo LJ, Nghiem PP, Kornegay JN. Short-term treatment of golden retriever muscular dystrophy (GRMD) dogs with rAAVrh74.MHCK7.GALGT2 induces muscle glycosylation and utrophin expression but has no significant effect on muscle strength. PLoS One 2021; 16:e0248721. [PMID: 33770101 PMCID: PMC7997012 DOI: 10.1371/journal.pone.0248721] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/03/2021] [Indexed: 11/30/2022] Open
Abstract
We have examined the effects of intravenous (IV) delivery of rAAVrh74.MHCK7.GALGT2 in the golden retriever muscular dystrophy (GRMD) model of Duchenne Muscular Dystrophy (DMD). After baseline testing, GRMD dogs were treated at 3 months of age and reassessed at 6 months. This 3–6 month age range is a period of rapid disease progression, thus offering a relatively short window to establish treatment efficacy. Measures analyzed included muscle AAV transduction, GALGT2 transgene expression, GALGT2-induced glycosylation, muscle pathology, and muscle function. A total of five dogs were treated, 4 at 2x1014vg/kg and one at 6x1014vgkg. The 2x1014vg/kg dose led to transduction of regions of the heart with 1–3 vector genomes (vg) per nucleus, while most skeletal muscles were transduced with 0.25–0.5vg/nucleus. GALGT2-induced glycosylation paralleled levels of myofiber vg transduction, with about 90% of cardiomyocytes having increased glycosylation versus 20–35% of all myofibers across the skeletal muscles tested. Conclusions from phenotypic testing were limited by the small number of dogs. Treated dogs had less pronounced fibrosis and overall lesion severity when compared to control groups, but surprisingly no significant changes in limb muscle function measures. GALGT2-treated skeletal muscle and heart had elevated levels of utrophin protein expression and GALGT2-induced expression of glycosylated α dystroglycan, providing further evidence of a treatment effect. Serum chemistry, hematology, and cardiac function measures were largely unchanged by treatment. Cumulatively, these data show that short-term intravenous treatment of GRMD dogs with rAAVrh74.MHCK7.GALGT2 at high doses can induce muscle glycosylation and utrophin expression and may be safe over a short 3-month interval, but that such treatments had only modest effects on muscle pathology and did not significantly improve muscle strength.
Collapse
Affiliation(s)
- Paul T. Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, United States of America
- * E-mail:
| | - Deborah A. Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States of America
| | - Sonia Hamilton
- Neuroscience Undergraduate Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Davin Packer
- Neuroscience Graduate Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Sharla M. Birch
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Amanda K. Bettis
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Cynthia J. Balog-Alvarez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Lee-Jae Guo
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Peter P. Nghiem
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| | - Joe N. Kornegay
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States of America
| |
Collapse
|
62
|
Fortunato F, Rossi R, Falzarano MS, Ferlini A. Innovative Therapeutic Approaches for Duchenne Muscular Dystrophy. J Clin Med 2021; 10:jcm10040820. [PMID: 33671409 PMCID: PMC7922390 DOI: 10.3390/jcm10040820] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common childhood muscular dystrophy affecting ~1:5000 live male births. Following the identification of pathogenic variations in the dystrophin gene in 1986, the underlining genotype/phenotype correlations emerged and the role of the dystrophin protein was elucidated in skeletal, smooth, and cardiac muscles, as well as in the brain. When the dystrophin protein is absent or quantitatively or qualitatively modified, the muscle cannot sustain the stress of repeated contractions. Dystrophin acts as a bridging and anchoring protein between the sarcomere and the sarcolemma, and its absence or reduction leads to severe muscle damage that eventually cannot be repaired, with its ultimate substitution by connective tissue and fat. The advances of an understanding of the molecular pathways affected in DMD have led to the development of many therapeutic strategies that tackle different aspects of disease etiopathogenesis, which have recently led to the first successful approved orphan drugs for this condition. The therapeutic advances in this field have progressed exponentially, with second-generation drugs now entering in clinical trials as gene therapy, potentially providing a further effective approach to the condition.
Collapse
|
63
|
Pedrazzani PS, Araújo TOP, Sigoli E, da Silva IR, da Roza DL, Chesca DL, Rassier DE, Cornachione AS. Twenty-one days of low-intensity eccentric training improve morphological characteristics and function of soleus muscles of mdx mice. Sci Rep 2021; 11:3579. [PMID: 33574358 PMCID: PMC7878734 DOI: 10.1038/s41598-020-79168-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/25/2020] [Indexed: 11/09/2022] Open
Abstract
Duchene muscular dystrophy (DMD) is caused by the absence of the protein dystrophin, which leads to muscle weakness, progressive degeneration, and eventually death due to respiratory failure. Low-intensity eccentric training (LIET) has been used as a rehabilitation method in skeletal muscles after disuse. Recently, LIET has also been used for rehabilitating dystrophic muscles, but its effects are still unclear. The purpose of this study was to investigate the effects of 21 days of LIET in dystrophic soleus muscle. Thirty-six male mdx mice were randomized into six groups (n = 6/each): mdx sedentary group; mdx training group-3 days; mdx training group-21 days; wild-type sedentary group; wild-type training group-3 days and wild-type training group-21 days. After the training sessions, animals were euthanized, and fragments of soleus muscles were removed for immunofluorescence and histological analyses, and measurements of active force and Ca2+ sensitivity of the contractile apparatus. Muscles of the mdx training group-21 days showed an improvement in morphological characteristics and an increase of active force when compared to the sedentary mdx group. The results show that LIET can improve the functionality of dystrophic soleus muscle in mice.
Collapse
Affiliation(s)
- Paulo S Pedrazzani
- Department of Physiological Science, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Tatiana O P Araújo
- Department of Physiological Science, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Emilly Sigoli
- Department of Physiological Science, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Isabella R da Silva
- Department of Physiological Science, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Daiane Leite da Roza
- Department of Neurosciences and Behaviour, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Deise Lucia Chesca
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dilson E Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, Canada
| | - Anabelle S Cornachione
- Department of Physiological Science, Federal University of São Carlos (UFSCar), São Carlos, Brazil.
| |
Collapse
|
64
|
Micheletto MLJ, Hermes TDA, Bertassoli BM, Petri G, Perez MM, Fonseca FLA, Carvalho AADS, Feder D. Ixazomib, an oral proteasome inhibitor, exhibits potential effect in dystrophin-deficient mdx mice. Int J Exp Pathol 2021; 102:11-21. [PMID: 33296126 PMCID: PMC7839951 DOI: 10.1111/iep.12383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/31/2020] [Accepted: 11/04/2020] [Indexed: 12/24/2022] Open
Abstract
Dystrophin deficiency makes the sarcolemma fragile and susceptible to degeneration in Duchenne muscular dystrophy. The proteasome is a multimeric protease complex and is central to the regulation of cellular proteins. Previous studies have shown that proteasome inhibition improved pathological changes in mdx mice. Ixazomib is the first oral proteasome inhibitor used as a therapy in multiple myeloma. This study investigated the effects of ixazomib on the dystrophic muscle of mdx mice. MDX mice were treated with ixazomib (7.5 mg/kg/wk by gavage) or 0.2 mL of saline for 12 weeks. The Kondziela test was performed to measure muscle strength. The tibialis anterior (TA) and diaphragm (DIA) muscles were used for morphological analysis, and blood samples were collected for biochemical measurement. We observed maintenance of the muscle strength in the animals treated with ixazomib. Treatment with ixazomib had no toxic effect on the mdx mouse. The morphological analysis showed a reduction in the inflammatory area and fibres with central nuclei in the TA and DIA muscles and an increase in the number of fibres with a diameter of 20 µm2 in the DIA muscle after treatment with ixazomib. There was an increase in the expression of dystrophin and utrophin in the TA and DIA muscles and a reduction in the expression of osteopontin and TGF-β in the DIA muscle of mdx mice treated with ixazomib. Ixazomib was thus shown to increase the expression of dystrophin and utrophin associated with improved pathological and functional changes in the dystrophic muscles of mdx mice.
Collapse
Affiliation(s)
| | - Tulio de Almeida Hermes
- Departament of Morphology and PhysiologyMedical Faculty of the ABCSanto AndréBrazil
- Departament of AnatomyFederal University of AlfenasAlfenasBrazil
| | | | - Giuliana Petri
- Departament of Morphology and PhysiologyMedical Faculty of the ABCSanto AndréBrazil
| | | | | | | | - David Feder
- Departament of Morphology and PhysiologyMedical Faculty of the ABCSanto AndréBrazil
| |
Collapse
|
65
|
Gibbs EM, McCourt JL, Shin KM, Hammond KG, Marshall JL, Crosbie RH. Loss of sarcospan exacerbates pathology in mdx mice, but does not affect utrophin amelioration of disease. Hum Mol Genet 2021; 30:149-159. [PMID: 33432327 PMCID: PMC8091037 DOI: 10.1093/hmg/ddaa264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/26/2020] [Accepted: 11/30/2020] [Indexed: 01/02/2023] Open
Abstract
The dystrophin-glycoprotein complex (DGC) is a membrane adhesion complex that provides structural stability at the sarcolemma by linking the myocyte's internal cytoskeleton and external extracellular matrix. In Duchenne muscular dystrophy (DMD), the absence of dystrophin leads to the loss of the DGC at the sarcolemma, resulting in sarcolemmal instability and progressive muscle damage. Utrophin (UTRN), an autosomal homolog of dystrophin, is upregulated in dystrophic muscle and partially compensates for the loss of dystrophin in muscle from patients with DMD. Here, we examine the interaction between Utr and sarcospan (SSPN), a small transmembrane protein that is a core component of both UTRN-glycoprotein complex (UGC) and DGC. We show that additional loss of SSPN causes an earlier onset of disease in dystrophin-deficient mdx mice by reducing the expression of the UGC at the sarcolemma. In order to further evaluate the role of SSPN in maintaining therapeutic levels of Utr at the sarcolemma, we tested the effect of Utr transgenic overexpression in mdx mice lacking SSPN (mdx:SSPN -/-:Utr-Tg). We found that overexpression of Utr restored SSPN to the sarcolemma in mdx muscle but that the ablation of SSPN in mdx muscle reduced Utr at the membrane. Nevertheless, Utr overexpression reduced central nucleation and improved grip strength in both lines. These findings demonstrate that high levels of Utr transgenic overexpression ameliorate the mdx phenotype independently of SSPN expression but that loss of SSPN may impair Utr-based mechanisms that rely on lower levels of Utr protein.
Collapse
Affiliation(s)
- Elizabeth M Gibbs
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Jackie L McCourt
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Kara M Shin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Katherine G Hammond
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Jamie L Marshall
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA.,Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.,Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
66
|
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: 47] [Impact Index Per Article: 15.7] [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
|
67
|
Chatzopoulou M, Emer E, Lecci C, Rowley JA, Casagrande AS, Moir L, Squire SE, Davies SG, Harriman S, Wynne GM, Wilson FX, Davies KE, Russell AJ. Decreasing HepG2 Cytotoxicity by Lowering the Lipophilicity of Benzo[d]oxazolephosphinate Ester Utrophin Modulators. ACS Med Chem Lett 2020; 11:2421-2427. [PMID: 33335663 DOI: 10.1021/acsmedchemlett.0c00405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022] Open
Abstract
Utrophin modulation is a disease-modifying therapeutic strategy for Duchenne muscular dystrophy that would be applicable to all patient populations. To improve the suboptimal profile of ezutromid, the first-in-class clinical candidate, a second generation of utrophin modulators bearing a phosphinate ester moiety was developed. This modification significantly improved the physicochemical and ADME properties, but one of the main lead molecules was found to have dose-limiting hepatotoxicity. In this work we describe how less lipophilic analogues retained utrophin modulatory activity in a reporter gene assay, upregulated utrophin protein in dystrophic mouse muscle cells, but also had improved physicochemical and ADME properties. Notably, ClogP was found to directly correlate with pIC50 in HepG2 cells, hence leading to a potentially safer toxicological profiles in this series. Compound 21 showed a balanced profile (H2K EC50: 4.17 μM, solubility: 477 μM, mouse hepatocyte T 1/2 > 240 min) and increased utrophin protein 1.6-fold in a Western blot assay.
Collapse
Affiliation(s)
- Maria Chatzopoulou
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
| | - Enrico Emer
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
| | - Cristina Lecci
- Evoetec (U.K.) Ltd, 114 Innovation Drive, Milton Park, Milton, Abingdon OX14 4RZ, U.K
| | - Jessica A. Rowley
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
| | | | - Lee Moir
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Sarah E. Squire
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Stephen G. Davies
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
| | - Shawn Harriman
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Graham M. Wynne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
| | - Francis X. Wilson
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Kay E. Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Angela J. Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, U.K
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3PQ, U.K
| |
Collapse
|
68
|
Sztretye M, Szabó L, Dobrosi N, Fodor J, Szentesi P, Almássy J, Magyar ZÉ, Dienes B, Csernoch L. From Mice to Humans: An Overview of the Potentials and Limitations of Current Transgenic Mouse Models of Major Muscular Dystrophies and Congenital Myopathies. Int J Mol Sci 2020; 21:ijms21238935. [PMID: 33255644 PMCID: PMC7728138 DOI: 10.3390/ijms21238935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Muscular dystrophies are a group of more than 160 different human neuromuscular disorders characterized by a progressive deterioration of muscle mass and strength. The causes, symptoms, age of onset, severity, and progression vary depending on the exact time point of diagnosis and the entity. Congenital myopathies are rare muscle diseases mostly present at birth that result from genetic defects. There are no known cures for congenital myopathies; however, recent advances in gene therapy are promising tools in providing treatment. This review gives an overview of the mouse models used to investigate the most common muscular dystrophies and congenital myopathies with emphasis on their potentials and limitations in respect to human applications.
Collapse
|
69
|
Lambert MR, Spinazzola JM, Widrick JJ, Pakula A, Conner JR, Chin JE, Owens JM, Kunkel LM. PDE10A Inhibition Reduces the Manifestation of Pathology in DMD Zebrafish and Represses the Genetic Modifier PITPNA. Mol Ther 2020; 29:1086-1101. [PMID: 33221436 PMCID: PMC7934586 DOI: 10.1016/j.ymthe.2020.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/04/2020] [Accepted: 11/15/2020] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder caused by mutations in the DMD gene. Absence of dystrophin protein leads to progressive degradation of skeletal and cardiac function and leads to premature death. Over the years, zebrafish have been increasingly used for studying DMD and are a powerful tool for drug discovery and therapeutic development. In our study, a birefringence screening assay led to identification of phosphodiesterase 10A (PDE10A) inhibitors that reduced the manifestation of dystrophic muscle phenotype in dystrophin-deficient sapje-like zebrafish larvae. PDE10A has been validated as a therapeutic target by pde10a morpholino-mediated reduction in muscle pathology and improvement in locomotion, muscle, and vascular function as well as long-term survival in sapje-like larvae. PDE10A inhibition in zebrafish and DMD patient-derived myoblasts were also associated with reduction of PITPNA expression that has been previously identified as a protective genetic modifier in two exceptional dystrophin-deficient golden retriever muscular dystrophy (GRMD) dogs that escaped the dystrophic phenotype. The combination of a phenotypic assay and relevant functional assessments in the sapje-like zebrafish enhances the potential for the prospective discovery of DMD therapeutics. Indeed, our results suggest a new application for a PDE10A inhibitor as a potential DMD therapeutic to be investigated in a mouse model of DMD.
Collapse
Affiliation(s)
- Matthias R Lambert
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Janelle M Spinazzola
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey J Widrick
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Anna Pakula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - James R Conner
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Janice E Chin
- Rare Disease Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Jane M Owens
- Rare Disease Research Unit, Pfizer, Cambridge, MA 02139, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; The Stem Cell Program, Boston Children's Hospital, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
70
|
Péladeau C, Jasmin BJ. Targeting IRES-dependent translation as a novel approach for treating Duchenne muscular dystrophy. RNA Biol 2020; 18:1238-1251. [PMID: 33164678 DOI: 10.1080/15476286.2020.1847894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Internal-ribosomal entry sites (IRES) are translational elements that allow the initiation machinery to start protein synthesis via internal initiation. IRESs promote tissue-specific translation in stress conditions when conventional cap-dependent translation is inhibited. Since many IRES-containing mRNAs are relevant to diseases, this cellular mechanism is emerging as an attractive therapeutic target for pharmacological and genetic modulations. Indeed, there has been growing interest over the past years in determining the therapeutic potential of IRESs for several disease conditions such as cancer, neurodegeneration and neuromuscular diseases including Duchenne muscular dystrophy (DMD). IRESs relevant for DMD have been identified in several transcripts whose protein product results in functional improvements in dystrophic muscles. Together, these converging lines of evidence indicate that activation of IRES-mediated translation of relevant transcripts in DMD muscle represents a novel and appropriate therapeutic strategy for DMD that warrants further investigation, particularly to identify agents that can modulate their activity.
Collapse
Affiliation(s)
- Christine Péladeau
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, and the Eric Poulin Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
71
|
A Role for Caveolin-3 in the Pathogenesis of Muscular Dystrophies. Int J Mol Sci 2020; 21:ijms21228736. [PMID: 33228026 PMCID: PMC7699313 DOI: 10.3390/ijms21228736] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Caveolae are the cholesterol-rich small invaginations of the plasma membrane present in many cell types including adipocytes, endothelial cells, epithelial cells, fibroblasts, smooth muscles, skeletal muscles and cardiac muscles. They serve as specialized platforms for many signaling molecules and regulate important cellular processes like energy metabolism, lipid metabolism, mitochondria homeostasis, and mechano-transduction. Caveolae can be internalized together with associated cargo. The caveolae-dependent endocytic pathway plays a role in the withdrawal of many plasma membrane components that can be sent for degradation or recycled back to the cell surface. Caveolae are formed by oligomerization of caveolin proteins. Caveolin-3 is a muscle-specific isoform, whose malfunction is associated with several diseases including diabetes, cancer, atherosclerosis, and cardiovascular diseases. Mutations in Caveolin-3 are known to cause muscular dystrophies that are collectively called caveolinopathies. Altered expression of Caveolin-3 is also observed in Duchenne’s muscular dystrophy, which is likely a part of the pathological process leading to muscle weakness. This review summarizes the major functions of Caveolin-3 in skeletal muscles and discusses its involvement in the pathology of muscular dystrophies.
Collapse
|
72
|
Banks GB, Chamberlain JS, Odom GL. Microutrophin expression in dystrophic mice displays myofiber type differences in therapeutic effects. PLoS Genet 2020; 16:e1009179. [PMID: 33175853 PMCID: PMC7682874 DOI: 10.1371/journal.pgen.1009179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/23/2020] [Accepted: 10/06/2020] [Indexed: 12/28/2022] Open
Abstract
Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (μUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-μUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for μUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed μUtrn. As such, we found that μUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike μUtrn, μdystrophin (μDys) expression did not show this fiber type preference. Interestingly, μUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & μUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.
Collapse
MESH Headings
- Animals
- Dependovirus/genetics
- Disease Models, Animal
- Dystrophin/genetics
- Gene Transfer Techniques
- Genetic Therapy/methods
- Genetic Vectors/genetics
- HEK293 Cells
- Humans
- Mice
- Mice, Inbred mdx
- Microscopy, Electron
- Muscle Contraction
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/ultrastructure
- Muscle, Skeletal
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/therapy
- Neuromuscular Junction/pathology
- Neuromuscular Junction/ultrastructure
- Sarcolemma/pathology
- Sarcolemma/ultrastructure
- Utrophin/genetics
- Utrophin/therapeutic use
Collapse
Affiliation(s)
- Glen B. Banks
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
| | - Jeffrey S. Chamberlain
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
- Department of BioChemistry, University of Washington, Seattle, Washington, United States of America
| | - Guy L. Odom
- Department of Neurology, University of Washington, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Wellstone Muscular Dystrophy Specialized Research Center, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
73
|
Łoboda A, Dulak J. Muscle and cardiac therapeutic strategies for Duchenne muscular dystrophy: past, present, and future. Pharmacol Rep 2020; 72:1227-1263. [PMID: 32691346 PMCID: PMC7550322 DOI: 10.1007/s43440-020-00134-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a severe X-linked neuromuscular childhood disorder that causes progressive muscle weakness and degeneration and results in functional decline, loss of ambulation and early death of young men due to cardiac or respiratory failure. Although the major cause of the disease has been known for many years-namely mutation in the DMD gene encoding dystrophin, one of the largest human genes-DMD is still incurable, and its treatment is challenging. METHODS A comprehensive and systematic review of literature on the gene, cell, and pharmacological experimental therapies aimed at restoring functional dystrophin or to counteract the associated processes contributing to disease progression like inflammation, fibrosis, calcium signaling or angiogenesis was carried out. RESULTS Although some therapies lead to satisfying effects in skeletal muscle, they are highly ineffective in the heart; therefore, targeting defective cardiac and respiratory systems is vital in DMD patients. Unfortunately, most of the pharmacological compounds treat only the symptoms of the disease. Some drugs addressing the underlying cause, like eteplirsen, golodirsen, and ataluren, have recently been conditionally approved; however, they can correct only specific mutations in the DMD gene and are therefore suitable for small sub-populations of affected individuals. CONCLUSION In this review, we summarize the possible therapeutic options and describe the current status of various, still imperfect, strategies used for attenuating the disease progression.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| |
Collapse
|
74
|
Yue F, Song C, Huang D, Narayanan N, Qiu J, Jia Z, Yuan Z, Oprescu SN, Roseguini BT, Deng M, Kuang S. PTEN Inhibition Ameliorates Muscle Degeneration and Improves Muscle Function in a Mouse Model of Duchenne Muscular Dystrophy. Mol Ther 2020; 29:132-148. [PMID: 33068545 DOI: 10.1016/j.ymthe.2020.09.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/10/2020] [Accepted: 09/20/2020] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by a mutation of the muscle membrane protein dystrophin and characterized by severe degeneration of myofibers, progressive muscle wasting, loss of mobility, and, ultimately, cardiorespiratory failure and premature death. Currently there is no cure for DMD. Herein, we report that skeletal muscle-specific knockout (KO) of the phosphatase and tensin homolog (Pten) gene in an animal model of DMD (mdx mice) alleviates myofiber degeneration and restores muscle function without increasing tumor incidence. Specifically, Pten KO normalizes myofiber size and prevents muscular atrophy, and it improves grip strength and exercise performance in mdx mice. Pten KO also reduces fibrosis and inflammation, and it ameliorates muscle pathology in mdx mice. Unbiased RNA sequencing reveals that Pten KO upregulates extracellular matrix and basement membrane components positively correlated with wound healing and suppresses negative regulators of wound healing and lipid biosynthesis, thus improving the integrity of muscle basement membrane at the ultrastructural level. Importantly, pharmacological inhibition of PTEN similarly ameliorates muscle pathology and improves muscle integrity and function in mdx mice. Our findings provide evidence that PTEN inhibition may represent a potential therapeutic strategy to restore muscle function in DMD.
Collapse
Affiliation(s)
- Feng Yue
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Changyou Song
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Di Huang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Jiamin Qiu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Zhihao Jia
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Zhengrong Yuan
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Stephanie N Oprescu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Bruno T Roseguini
- Department of Health and Kinesiology, Purdue University, West Lafayette, IN 47907, USA
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
75
|
Shu C, Parfenova L, Mokhonova E, Collado JR, Damoiseaux R, Campagna J, John V, Crosbie RH. High-throughput screening identifies modulators of sarcospan that stabilize muscle cells and exhibit activity in the mouse model of Duchenne muscular dystrophy. Skelet Muscle 2020; 10:26. [PMID: 32948250 PMCID: PMC7499884 DOI: 10.1186/s13395-020-00244-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/01/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a degenerative muscle disease caused by mutations in the dystrophin gene. Loss of dystrophin prevents the formation of a critical connection between the muscle cell membrane and the extracellular matrix. Overexpression of sarcospan (SSPN) in the mouse model of DMD restores the membrane connection and reduces disease severity, making SSPN a promising therapeutic target for pharmacological upregulation. METHODS Using a previously described cell-based promoter reporter assay of SSPN gene expression (hSSPN-EGFP), we conducted high-throughput screening on libraries of over 200,000 curated small molecules to identify SSPN modulators. The hits were validated in both hSSPN-EGFP and hSSPN-luciferase reporter cells. Hit selection was conducted on dystrophin-deficient mouse and human myotubes with assessments of (1) SSPN gene expression using quantitative PCR and (2) SSPN protein expression using immunoblotting and an ELISA. A membrane stability assay using osmotic shock was used to validate the functional effects of treatment followed by cell surface biotinylation to label cell surface proteins. Dystrophin-deficient mdx mice were treated with compound, and muscle was subjected to quantitative PCR to assess SSPN gene expression. RESULTS We identified and validated lead compounds that increased SSPN gene and protein expression in dystrophin-deficient mouse and human muscle cells. The lead compound OT-9 increased cell membrane localization of compensatory laminin-binding adhesion complexes and improved membrane stability in DMD myotubes. We demonstrated that the membrane stabilizing benefit is dependent on SSPN. Intramuscular injection of OT-9 in the mouse model of DMD increased SSPN gene expression. CONCLUSIONS This study identifies a pharmacological approach to treat DMD and sets the path for the development of SSPN-based therapies.
Collapse
Affiliation(s)
- Cynthia Shu
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
- Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, CA, USA
| | - Liubov Parfenova
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Ekaterina Mokhonova
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
- Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, CA, USA
| | - Judd R Collado
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Department of Molecular and Medicinal Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Jesus Campagna
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
- Drug Discovery Lab, University of California Los Angeles, Los Angeles, CA, USA
| | - Varghese John
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
- Drug Discovery Lab, University of California Los Angeles, Los Angeles, CA, USA
| | - Rachelle H Crosbie
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
- Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
| |
Collapse
|
76
|
Lovering RM, Iyer SR, Edwards B, Davies KE. Alterations of neuromuscular junctions in Duchenne muscular dystrophy. Neurosci Lett 2020; 737:135304. [PMID: 32818587 DOI: 10.1016/j.neulet.2020.135304] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/09/2020] [Accepted: 08/11/2020] [Indexed: 12/18/2022]
Abstract
The focus of this review is on Duchenne muscular dystrophy (DMD), which is caused by the absence of the protein dystrophin and is characterized as a neuromuscular disease in which muscle weakness, increased susceptibility to muscle injury, and inadequate repair appear to underlie the pathology. Considerable attention has been dedicated to studying muscle fiber damage, but data show that both human patients and animal models for DMD present with fragmented neuromuscular junction (NMJ) morphology. In addition to pre- and post-synaptic abnormalities, studies indicate increased susceptibility of the NMJ to contraction-induced injury, with corresponding functional changes in neuromuscular transmission and nerve-evoked electromyographic activity. Such findings suggest that alterations in the NMJ of dystrophic muscle may play a role in muscle weakness via impairment of neuromuscular transmission. Further work is needed to fully understand the role of the NMJ in the weakness, susceptibility to injury, and progressive wasting associated with DMD.
Collapse
Affiliation(s)
- Richard M Lovering
- Departments of Orthopaedics and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA; University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Shama R Iyer
- Departments of Orthopaedics and Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Benjamin Edwards
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Kay E Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
77
|
Babbs A, Berg A, Chatzopoulou M, Davies KE, Davies SG, Edwards B, Elsey DJ, Emer E, Guiraud S, Harriman S, Lecci C, Moir L, Peters D, Robinson N, Rowley JA, Russell AJ, Squire SE, Tinsley JM, Wilson FX, Wynne GM. 2-Arylbenzo[ d]oxazole Phosphinate Esters as Second-Generation Modulators of Utrophin for the Treatment of Duchenne Muscular Dystrophy. J Med Chem 2020; 63:7880-7891. [PMID: 32551645 DOI: 10.1021/acs.jmedchem.0c00807] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Utrophin modulation is a promising therapeutic strategy for Duchenne muscular dystrophy (DMD), which should be applicable to all patient populations. Following on from ezutromid, the first-generation utrophin modulator, we describe the development of a second generation of utrophin modulators, based on the bioisosteric replacement of the sulfone group with a phosphinate ester and substitution of the metabolically labile naphthalene with a haloaryl substituent. The improved physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties, further reflected in the enhanced pharmacokinetic profile of the most advanced compounds, 30 and 27, led to significantly better in vivo exposure compared to ezutromid and alleviation of the dystrophic phenotype in mdx mice. While 30 was found to have dose-limiting hepatotoxicity, 27 and its enantiomers exhibited limited off-target effects, resulting in a safe profile and highlighting their potential utility as next-generation utrophin modulators suitable for progression toward a future DMD therapy.
Collapse
Affiliation(s)
- Arran Babbs
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Adam Berg
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Maria Chatzopoulou
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Kay E Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Stephen G Davies
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Benjamin Edwards
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - David J Elsey
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Enrico Emer
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Simon Guiraud
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Shawn Harriman
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Cristina Lecci
- Evotec (UK) Ltd, 114 Innovation Dr, Milton Park, Milton, Abingdon OX14 4RZ, U.K
| | - Lee Moir
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - David Peters
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Neil Robinson
- S.H.B. Enterprises Ltd, 55 Station Road, Beaconsfield HP19 1QL, U.K
| | - Jessica A Rowley
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.,Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3PQ, U.K
| | - Sarah E Squire
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, U.K
| | - Jonathon M Tinsley
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Francis X Wilson
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon, Oxfordshire OX14 4SB, U.K
| | - Graham M Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K
| |
Collapse
|
78
|
Datta N, Ghosh PS. Update on Muscular Dystrophies with Focus on Novel Treatments and Biomarkers. Curr Neurol Neurosci Rep 2020; 20:14. [DOI: 10.1007/s11910-020-01034-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
79
|
Heterogenetic parabiosis between healthy and dystrophic mice improve the histopathology in muscular dystrophy. Sci Rep 2020; 10:7075. [PMID: 32341395 PMCID: PMC7184587 DOI: 10.1038/s41598-020-64042-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/09/2020] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease, characterized by mutations in the X-linked dystrophin, that has several therapeutic options but no curative treatment. Transplantation of muscle progenitor cells for treatment of DMD has been widely investigated; however, its application is hindered by limited cell survival due to the harmful dystrophic microenvironment. An alternative approach to utilize progenitor cells and circulatory factors and to improve the dystrophic muscle pathology and microenvironment is through parabiotic pairing, where mice are surgically sutured to create a joint circulatory system. Parabiotic mice were generated by surgically joining wild type (WT) mice expressing green fluorescent protein (GFP) with mdx mice. These mice developed a common circulation (approximately 50% green cells in the blood of mdx mice) 2-weeks after parabiotic pairing. We observed significantly improved dystrophic muscle pathology, including decreased inflammation, necrotic fibers and fibrosis in heterogenetic parabionts. Importantly, the GFP + cells isolated from the mdx mice (paired with GFP mice) underwent myogenic differentiation in vitro and expressed markers of mesenchymal stem cells and macrophages, which may potentially be involved in the improvement of dystrophic muscle pathology. These observations suggest that changing the dystrophic microenvironment can be a new approach to treat DMD.
Collapse
|
80
|
Péladeau C, Adam N, Bronicki LM, Coriati A, Thabet M, Al-Rewashdy H, Vanstone J, Mears A, Renaud JM, Holcik M, Jasmin BJ. Identification of therapeutics that target eEF1A2 and upregulate utrophin A translation in dystrophic muscles. Nat Commun 2020; 11:1990. [PMID: 32332749 PMCID: PMC7181625 DOI: 10.1038/s41467-020-15971-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 04/06/2020] [Indexed: 01/10/2023] Open
Abstract
Up-regulation of utrophin in muscles represents a promising therapeutic strategy for the treatment of Duchenne Muscular Dystrophy. We previously demonstrated that eEF1A2 associates with the 5’UTR of utrophin A to promote IRES-dependent translation. Here, we examine whether eEF1A2 directly regulates utrophin A expression and identify via an ELISA-based high-throughput screen, FDA-approved drugs that upregulate both eEF1A2 and utrophin A. Our results show that transient overexpression of eEF1A2 in mouse muscles causes an increase in IRES-mediated translation of utrophin A. Through the assessment of our screen, we reveal 7 classes of FDA-approved drugs that increase eEF1A2 and utrophin A protein levels. Treatment of mdx mice with the 2 top leads results in multiple improvements of the dystrophic phenotype. Here, we report that IRES-mediated translation of utrophin A via eEF1A2 is a critical mechanism of regulating utrophin A expression and reveal the potential of repurposed drugs for treating DMD via this pathway. One potential approach for the treatment of Duchenne muscular dysrophy is to increase expression of the dystrophin homolog utrophin. Here, the authors show that eEF1A2 regulates utrophin expression, and show that 2 FDA-approved drugs upregulate eEIF1A2 and utrophin level in mice, leading to improvement of the dystrophic phenotype.
Collapse
Affiliation(s)
- Christine Péladeau
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.,Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Nadine Adam
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.,Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Lucas M Bronicki
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.,Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Adèle Coriati
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Mohamed Thabet
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Hasanen Al-Rewashdy
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.,Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Jason Vanstone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 5B2, Canada
| | - Alan Mears
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 5B2, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Martin Holcik
- Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada. .,Centre for Neuromuscular Disease, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
81
|
van Westering TLE, Lomonosova Y, Coenen-Stass AML, Betts CA, Bhomra A, Hulsker M, Clark LE, McClorey G, Aartsma-Rus A, van Putten M, Wood MJA, Roberts TC. Uniform sarcolemmal dystrophin expression is required to prevent extracellular microRNA release and improve dystrophic pathology. J Cachexia Sarcopenia Muscle 2020; 11:578-593. [PMID: 31849191 PMCID: PMC7113513 DOI: 10.1002/jcsm.12506] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 09/25/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disorder caused by genetic loss of dystrophin protein. Extracellular microRNAs (ex-miRNAs) are putative, minimally invasive biomarkers of DMD. Specific ex-miRNAs (e.g. miR-1, miR-133a, miR-206, and miR-483) are highly up-regulated in the serum of DMD patients and dystrophic animal models and are restored to wild-type levels following exon skipping-mediated dystrophin rescue in mdx mice. As such, ex-miRNAs are promising pharmacodynamic biomarkers of exon skipping efficacy. Here, we aimed to determine the degree to which ex-miRNA levels reflect the underlying level of dystrophin protein expression in dystrophic muscle. METHODS Candidate ex-miRNA biomarker levels were investigated in mdx mice in which dystrophin was restored with peptide-PMO (PPMO) exon skipping conjugates and in mdx-XistΔhs mice that express variable amounts of dystrophin from birth as a consequence of skewed X-chromosome inactivation. miRNA profiling was performed in mdx-XistΔhs mice using the FirePlex methodology and key results validated by small RNA TaqMan RT-qPCR. The muscles from each animal model were further characterized by dystrophin western blot and immunofluorescence staining. RESULTS The restoration of ex-myomiR abundance observed following PPMO treatment was not recapitulated in the high dystrophin-expressing mdx-XistΔhs group, despite these animals expressing similar amounts of total dystrophin protein (~37% of wild-type levels). Instead, ex-miRNAs were present at high levels in mdx-XistΔhs mice regardless of dystrophin expression. PPMO-treated muscles exhibited a uniform pattern of dystrophin localization and were devoid of regenerating fibres, whereas mdx-XistΔhs muscles showed non-homogeneous dystrophin staining and sporadic regenerating foci. CONCLUSIONS Uniform dystrophin expression is required to prevent ex-miRNA release, stabilize myofiber turnover, and attenuate pathology in dystrophic muscle.
Collapse
Affiliation(s)
- Tirsa L E van Westering
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Yulia Lomonosova
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Anna M L Coenen-Stass
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK
| | - Corinne A Betts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Amarjit Bhomra
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Margriet Hulsker
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Lucy E Clark
- Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Graham McClorey
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, UK.,Department of Paediatrics, University of Oxford, South Parks Road, Oxford, UK.,Sanford Burnham Prebys Medical Discovery Institute, Development, Aging and Regeneration Program, La Jolla, CA, USA
| |
Collapse
|
82
|
Hofmann I, Kemter E, Theobalt N, Fiedler S, Bidlingmaier M, Hinrichs A, Aichler M, Burkhardt K, Klymiuk N, Wolf E, Wanke R, Blutke A. Linkage between growth retardation and pituitary cell morphology in a dystrophin-deficient pig model of Duchenne muscular dystrophy. Growth Horm IGF Res 2020; 51:6-16. [PMID: 31926372 DOI: 10.1016/j.ghir.2019.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Human patients with Duchenne muscular dystrophy (DMD) commonly exhibit a short stature, but the pathogenesis of this growth retardation is not completely understood. Due to the suspected involvement of the growth hormone/insulin-like growth factor 1 (GH/IGF1) system, controversial therapeutic approaches have been developed, including both GH- administration, as well as GH-inhibition. In the present study, we examined relevant histomorphological and ultrastructural features of adenohypophyseal GH-producing somatotroph cells in a porcine DMD model. METHODS The numbers and volumes of immunohistochemically labelled somatotroph cells were determined in consecutive semi-thin sections of plastic resin embedded adenohypophyseal tissue samples using unbiased state-of-the-art quantitative stereological analysis methods. RESULTS DMD pigs displayed a significant growth retardation, accounting for a 55% reduction of body weight, accompanied by a significant 50% reduction of the number of somatotroph cells, as compared to controls. However, the mean volumes of somatotroph cells and the volume of GH-granules per cell were not altered. Western blot analyses of the adenohypophyseal protein samples showed no differences in the relative adenohypophyseal GH-abundance between DMD pigs and controls. CONCLUSION The findings of this study do not provide evidence for involvement of somatotroph cells in the pathogenesis of growth retardation of DMD pigs. These results are in contrast with previous findings in other dystrophin-deficient animal models, such as the golden retriever model of Duchenne muscular dystrophy, where increased mean somatotroph cell volumes and elevated volumes of intracellular GH-granules were reported and associated with DMD-related growth retardation. Possible reasons for the differences of somatotroph morphology observed in different DMD models are discussed.
Collapse
Affiliation(s)
- I Hofmann
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - E Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany; Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - N Theobalt
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - S Fiedler
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - M Bidlingmaier
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - A Hinrichs
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany; Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - M Aichler
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - K Burkhardt
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany; Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - N Klymiuk
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany; Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleißheim, Germany
| | - E Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Centre and Department of Veterinary Sciences, Ludwig-Maximilians-Universität München, Munich, Germany; Centre for Innovative Medical Models (CiMM), Ludwig-Maximilians-Universität München, Oberschleißheim, Germany; Laboratory for Functional Genome Analysis (LAFUGA), Gene Centre, Ludwig-Maximilians-Universität München, Munich, Germany
| | - R Wanke
- Institute of Veterinary Pathology at the Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - A Blutke
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany.
| |
Collapse
|
83
|
Razzoli M, Lindsay A, Law ML, Chamberlain CM, Southern WM, Berg M, Osborn J, Engeland WC, Metzger JM, Ervasti JM, Bartolomucci A. Social stress is lethal in the mdx model of Duchenne muscular dystrophy. EBioMedicine 2020; 55:102700. [PMID: 32192914 PMCID: PMC7251247 DOI: 10.1016/j.ebiom.2020.102700] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 02/10/2020] [Accepted: 02/18/2020] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is caused by the loss of dystrophin. Severe and ultimately lethal, DMD progresses relatively slowly in that patients become wheelchair bound only around age twelve with a survival expectancy reaching the third decade of life. Methods The mildly-affected mdx mouse model of DMD, and transgenic DysΔMTB-mdx and Fiona-mdx mice expressing dystrophin or utrophin, respectively, were exposed to either mild (scruffing) or severe (subordination stress) stress paradigms and profiled for their behavioral and physiological responses. A subgroup of mdx mice exposed to subordination stress were pretreated with the beta-blocker metoprolol. Findings Subordination stress caused lethality in ∼30% of mdx mice within 24 h and ∼70% lethality within 48 h, which was not rescued by metoprolol. Lethality was associated with heart damage, waddling gait and hypo-locomotion, as well as marked up-regulation of the hypothalamus-pituitary-adrenocortical axis. A novel cardiovascular phenotype emerged in mdx mice, in that scruffing caused a transient drop in arterial pressure, while subordination stress caused severe and sustained hypotension with concurrent tachycardia. Transgenic expression of dystrophin or utrophin in skeletal muscle protected mdx mice from scruffing and social stress-induced responses including mortality. Interpretation We have identified a robust new stress phenotype in the otherwise mildly affected mdx mouse that suggests relatively benign handling may impact the outcome of behavioural experiments, but which should also expedite the knowledge-based therapy development for DMD. Funding Greg Marzolf Jr. Foundation, Summer's Wish Fund, NIAMS, Muscular Dystrophy Association, University of Minnesota and John and Cheri Gunvalson Trust.
Collapse
Affiliation(s)
- Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Angus Lindsay
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Michelle L Law
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Christopher M Chamberlain
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William M Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Madeleine Berg
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - John Osborn
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - William C Engeland
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States.
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States.
| |
Collapse
|
84
|
Kanamori H, Naruse G, Yoshida A, Minatoguchi S, Watanabe T, Kawaguchi T, Yamada Y, Mikami A, Kawasaki M, Takemura G, Minatoguchi S. Metformin Enhances Autophagy and Provides Cardioprotection in δ-Sarcoglycan Deficiency-Induced Dilated Cardiomyopathy. Circ Heart Fail 2020; 12:e005418. [PMID: 30922066 DOI: 10.1161/circheartfailure.118.005418] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Metformin is a popular antidiabetic agent that is also used to treat heart failure patients with type 2 diabetes mellitus. Several reports suggest that metformin may also have cardioprotective effects in patients without diabetes mellitus. In the present study, we investigated the possible therapeutic effect of metformin in heart failure and its underlying molecular mechanisms using a δ-sarcoglycan-deficient mouse model of dilated cardiomyopathy. METHODS AND RESULTS Thirty-two-week-old δ-sarcoglycan-deficient mice exhibiting established cardiomyopathy with extensive left ventricular dilatation and dysfunction were administered saline or metformin (200 mg/kg per day) for 4 weeks using osmotic mini-pumps. Metformin partially reversed the left ventricular dilatation (reverse remodeling) and significantly improved cardiac function. The hearts of metformin-treated mice showed less fibrosis, less cardiomyocyte hypertrophy, and fewer degenerative subcellular changes than saline-treated mice. These effects were accompanied by restored expression of the sarcomeric proteins myosin heavy chain and troponin I, and their transcription factor, GATA-4. Autophagy was enhanced in the hearts from metformin-treated mice, as indicated by increase of myocardial microtubule-associated protein-1 LC-3 (light chain 3)-II levels and LC3-II/-I ratios as well as levels of cathepsin D and ATP. In addition, increased numbers of autophagic vacuoles and lysosomes were accompanied increased AMP-activated protein kinase activity and suppression of mammalian target of rapamycin phosphorylation. Finally, autophagic flux assays using short-term chloroquine treatment revealed that autophagy was activated in δ-sarcoglycan-deficient hearts and was further augmented by metformin treatment. CONCLUSIONS Metformin is a beneficial pharmacological tool that mitigates heart failure caused by δ-sarcoglycan deficiency in association with enhanced autophagy.
Collapse
Affiliation(s)
- Hiromitsu Kanamori
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Genki Naruse
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Akihiro Yoshida
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Shingo Minatoguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Takatomo Watanabe
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Tomonori Kawaguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Yoshihisa Yamada
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Atsushi Mikami
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Masanori Kawasaki
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| | - Genzou Takemura
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan.,Department of Internal Medicine, Asahi University, Mizuho, Japan (G.T.)
| | - Shinya Minatoguchi
- Department of Cardiology (H.K., G.N., A.Y., S.M., T.W., T.K., Y.Y., A.M., M.K., G.T., S.M.), Gifu University Graduate School of Medicine, Japan
| |
Collapse
|
85
|
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: 10] [Impact Index Per Article: 2.5] [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
|
86
|
High-throughput identification of post-transcriptional utrophin up-regulators for Duchenne muscle dystrophy (DMD) therapy. Sci Rep 2020; 10:2132. [PMID: 32034254 PMCID: PMC7005813 DOI: 10.1038/s41598-020-58737-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/06/2019] [Indexed: 12/31/2022] Open
Abstract
Upregulation of endogenous utrophin offers great promise for treating DMD, as it can functionally compensate for the lack of dystrophin caused by DMD gene mutations, without the immunogenic concerns associated with delivering dystrophin. However, post-transcriptional repression mechanisms targeting the 5′ and 3′ untranslated regions (UTRs) of utrophin mRNA significantly limit the magnitude of utrophin upregulation achievable by promoter activation. Using a utrophin 5′3′UTR reporter assay, we performed a high-throughput screen (HTS) for small molecules capable of relieving utrophin post-transcriptional repression. We identified 27 hits that were ranked using a using an algorithm that we designed for hit prioritization that we call Hit to Lead Prioritization Score (H2LPS). The top 10 hits were validated using an orthogonal assay for endogenous utrophin expression. Evaluation of the top scoring hit, Trichostatin A (TSA), demonstrated utrophin upregulation and functional improvement in the mdx mouse model of DMD. TSA and the other small molecules identified here represent potential starting points for DMD drug discovery efforts.
Collapse
|
87
|
Wilkinson IVL, Perkins KJ, Dugdale H, Moir L, Vuorinen A, Chatzopoulou M, Squire SE, Monecke S, Lomow A, Geese M, Charles PD, Burch P, Tinsley JM, Wynne GM, Davies SG, Wilson FX, Rastinejad F, Mohammed S, Davies KE, Russell AJ. Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201912392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Isabel V. L. Wilkinson
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Kelly J. Perkins
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Hannah Dugdale
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Lee Moir
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Aini Vuorinen
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Maria Chatzopoulou
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Sarah E. Squire
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Sebastian Monecke
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Alexander Lomow
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Marcus Geese
- Evotec International GmbHManfred Eigen Campus Essener Bogen 7 22419 Hamburg Germany
| | - Philip D. Charles
- Department of BiochemistryUniversity of Oxford South Parks Rd Oxford OX1 3QU UK
- Target Discovery InstituteUniversity of OxfordOld Road Campus Roosevelt Drive Oxford OX3 7FZ UK
| | - Peter Burch
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Jonathan M. Tinsley
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Graham M. Wynne
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Stephen G. Davies
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
| | - Francis X. Wilson
- Summit Therapeutics plc. 136a Eastern Avenue, Milton Park Abingdon Oxfordshire OX14 4SB UK
| | - Fraydoon Rastinejad
- Target Discovery InstituteUniversity of OxfordOld Road Campus Roosevelt Drive Oxford OX3 7FZ UK
| | - Shabaz Mohammed
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
- Department of BiochemistryUniversity of Oxford South Parks Rd Oxford OX1 3QU UK
| | - Kay E. Davies
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene Function South Parks Road Oxford OX1 3PT UK
| | - Angela J. Russell
- Department of ChemistryUniversity of OxfordChemistry Research Laboratory Mansfield Road Oxford OX1 3TA UK
- Department of PharmacologyUniversity of Oxford Mansfield Road Oxford OX1 3PQ UK
| |
Collapse
|
88
|
Wilkinson IVL, Perkins KJ, Dugdale H, Moir L, Vuorinen A, Chatzopoulou M, Squire SE, Monecke S, Lomow A, Geese M, Charles PD, Burch P, Tinsley JM, Wynne GM, Davies SG, Wilson FX, Rastinejad F, Mohammed S, Davies KE, Russell AJ. Chemical Proteomics and Phenotypic Profiling Identifies the Aryl Hydrocarbon Receptor as a Molecular Target of the Utrophin Modulator Ezutromid. Angew Chem Int Ed Engl 2020; 59:2420-2428. [PMID: 31755636 PMCID: PMC7003794 DOI: 10.1002/anie.201912392] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/13/2019] [Indexed: 12/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal muscle-wasting disease arising from mutations in the dystrophin gene. Upregulation of utrophin to compensate for the missing dystrophin offers a potential therapy independent of patient genotype. The first-in-class utrophin modulator ezutromid/SMT C1100 was developed from a phenotypic screen through to a Phase 2 clinical trial. Promising efficacy and evidence of target engagement was observed in DMD patients after 24 weeks of treatment, however trial endpoints were not met after 48 weeks. The objective of this study was to understand the mechanism of action of ezutromid which could explain the lack of sustained efficacy and help development of new generations of utrophin modulators. Using chemical proteomics and phenotypic profiling we show that the aryl hydrocarbon receptor (AhR) is a target of ezutromid. Several lines of evidence demonstrate that ezutromid binds AhR with an apparent KD of 50 nm and behaves as an AhR antagonist. Furthermore, other reported AhR antagonists also upregulate utrophin, showing that this pathway, which is currently being explored in other clinical applications including oncology and rheumatoid arthritis, could also be exploited in future DMD therapies.
Collapse
Affiliation(s)
- Isabel V. L. Wilkinson
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Kelly J. Perkins
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Hannah Dugdale
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Lee Moir
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Aini Vuorinen
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Maria Chatzopoulou
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Sarah E. Squire
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Sebastian Monecke
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Alexander Lomow
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Marcus Geese
- Evotec International GmbHManfred Eigen CampusEssener Bogen 722419HamburgGermany
| | - Philip D. Charles
- Department of BiochemistryUniversity of OxfordSouth Parks RdOxfordOX1 3QUUK
- Target Discovery InstituteUniversity of OxfordOld Road CampusRoosevelt DriveOxfordOX3 7FZUK
| | - Peter Burch
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Jonathan M. Tinsley
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Graham M. Wynne
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Stephen G. Davies
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
| | - Francis X. Wilson
- Summit Therapeutics plc.136a Eastern Avenue, Milton ParkAbingdonOxfordshireOX14 4SBUK
| | - Fraydoon Rastinejad
- Target Discovery InstituteUniversity of OxfordOld Road CampusRoosevelt DriveOxfordOX3 7FZUK
| | - Shabaz Mohammed
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
- Department of BiochemistryUniversity of OxfordSouth Parks RdOxfordOX1 3QUUK
| | - Kay E. Davies
- Department of Physiology, Anatomy and GeneticsUniversity of OxfordSir Henry Wellcome Building of Gene FunctionSouth Parks RoadOxfordOX1 3PTUK
| | - Angela J. Russell
- Department of ChemistryUniversity of OxfordChemistry Research LaboratoryMansfield RoadOxfordOX1 3TAUK
- Department of PharmacologyUniversity of OxfordMansfield RoadOxfordOX1 3PQUK
| |
Collapse
|
89
|
Lindsay A, Baumann CW, Rebbeck RT, Yuen SL, Southern WM, Hodges JS, Cornea RL, Thomas DD, Ervasti JM, Lowe DA. Mechanical factors tune the sensitivity of mdx muscle to eccentric strength loss and its protection by antioxidant and calcium modulators. Skelet Muscle 2020; 10:3. [PMID: 32007101 PMCID: PMC6995146 DOI: 10.1186/s13395-020-0221-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/09/2020] [Indexed: 12/17/2022] Open
Abstract
Background Dystrophin deficiency sensitizes skeletal muscle of mice to eccentric contraction (ECC)-induced strength loss. ECC protocols distinguish dystrophin-deficient from healthy, wild type muscle, and test the efficacy of therapeutics for Duchenne muscular dystrophy (DMD). However, given the large lab-to-lab variability in ECC-induced strength loss of dystrophin-deficient mouse skeletal muscle (10–95%), mechanical factors of the contraction likely impact the degree of loss. Therefore, the purpose of this study was to evaluate the extent to which mechanical variables impact sensitivity of dystrophin-deficient mouse skeletal muscle to ECC. Methods We completed ex vivo and in vivo muscle preparations of the dystrophin-deficient mdx mouse and designed ECC protocols within physiological ranges of contractile parameters (length change, velocity, contraction duration, and stimulation frequencies). To determine whether these contractile parameters affected known factors associated with ECC-induced strength loss, we measured sarcolemmal damage after ECC as well as strength loss in the presence of the antioxidant N-acetylcysteine (NAC) and small molecule calcium modulators that increase SERCA activity (DS-11966966 and CDN1163) or lower calcium leak from the ryanodine receptor (Chloroxine and Myricetin). Results The magnitude of length change, work, and stimulation duration ex vivo and in vivo of an ECC were the most important determinants of strength loss in mdx muscle. Passive lengthening and submaximal stimulations did not induce strength loss. We further showed that sarcolemmal permeability was associated with muscle length change, but it only accounted for a minimal fraction (21%) of the total strength loss (70%). The magnitude of length change also significantly influenced the degree to which NAC and small molecule calcium modulators protected against ECC-induced strength loss. Conclusions These results indicate that ECC-induced strength loss of mdx skeletal muscle is dependent on the mechanical properties of the contraction and that mdx muscle is insensitive to ECC at submaximal stimulation frequencies. Rigorous design of ECC protocols is critical for effective use of strength loss as a readout in evaluating potential therapeutics for muscular dystrophy.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA.,Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA.,Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, VIC, 3220, Australia
| | - Cory W Baumann
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA
| | - Robyn T Rebbeck
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - Samantha L Yuen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - William M Southern
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - James S Hodges
- Division of Biostatistics, University of Minnesota, A460 Mayo Building, 420 Delaware Street SE, Minneapolis, 55455, USA
| | - Razvan L Cornea
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - David D Thomas
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, 55455, USA
| | - Dawn A Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, MMC 388, 420 Delaware Street SE, Minneapolis, 55455, USA.
| |
Collapse
|
90
|
Shu C, Kaxon-Rupp AN, Collado JR, Damoiseaux R, Crosbie RH. Development of a high-throughput screen to identify small molecule enhancers of sarcospan for the treatment of Duchenne muscular dystrophy. Skelet Muscle 2019; 9:32. [PMID: 31831063 PMCID: PMC6907331 DOI: 10.1186/s13395-019-0218-x] [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] [Received: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is caused by loss of sarcolemma connection to the extracellular matrix. Transgenic overexpression of the transmembrane protein sarcospan (SSPN) in the DMD mdx mouse model significantly reduces disease pathology by restoring membrane adhesion. Identifying SSPN-based therapies has the potential to benefit patients with DMD and other forms of muscular dystrophies caused by deficits in muscle cell adhesion. METHODS Standard cloning methods were used to generate C2C12 myoblasts stably transfected with a fluorescence reporter for human SSPN promoter activity. Assay development and screening were performed in a core facility using liquid handlers and imaging systems specialized for use with a 384-well microplate format. Drug-treated cells were analyzed for target gene expression using quantitative PCR and target protein expression using immunoblotting. RESULTS We investigated the gene expression profiles of SSPN and its associated proteins during myoblast differentiation into myotubes, revealing an increase in expression after 3 days of differentiation. We created C2C12 muscle cells expressing an EGFP reporter for SSPN promoter activity and observed a comparable increase in reporter levels during differentiation. Assay conditions for high-throughput screening were optimized for a 384-well microplate format and a high-content imager for the visualization of reporter levels. We conducted a screen of 3200 compounds and identified seven hits, which include an overrepresentation of L-type calcium channel antagonists, suggesting that SSPN gene activity is sensitive to calcium. Further validation of a select hit revealed that the calcium channel inhibitor felodipine increased SSPN transcript and protein levels in both wild-type and dystrophin-deficient myotubes, without increasing differentiation. CONCLUSIONS We developed a stable muscle cell line containing the promoter region of the human SSPN protein fused to a fluorescent reporter. Using the reporter cells, we created and validated a scalable, cell-based assay that is able to identify compounds that increase SSPN promoter reporter, transcript, and protein levels in wild-type and dystrophin-deficient muscle cells.
Collapse
Affiliation(s)
- Cynthia Shu
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.,Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, USA
| | - Ariana N Kaxon-Rupp
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Judd R Collado
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Department of Molecular and Medicinal Pharmacology, University of California Los Angeles, Los Angeles, USA.,California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Rachelle H Crosbie
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA. .,Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA. .,Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, USA. .,Department of Neurology David Geffen School of Medicine, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
| |
Collapse
|
91
|
Chatzopoulou M, Claridge TDW, Davies KE, Davies SG, Elsey DJ, Emer E, Fletcher AM, Harriman S, Robinson N, Rowley JA, Russell AJ, Tinsley JM, Weaver R, Wilkinson IVL, Willis NJ, Wilson FX, Wynne GM. Isolation, Structural Identification, Synthesis, and Pharmacological Profiling of 1,2-trans-Dihydro-1,2-diol Metabolites of the Utrophin Modulator Ezutromid. J Med Chem 2019; 63:2547-2556. [DOI: 10.1021/acs.jmedchem.9b01547] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Maria Chatzopoulou
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Tim D. W. Claridge
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Kay E. Davies
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Stephen G. Davies
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - David J. Elsey
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon OX14 4SB, United Kingdom
| | - Enrico Emer
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Ai M. Fletcher
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Shawn Harriman
- Summit Therapeutics plc, One Broadway, 14th Floor, Cambridge, Massachusetts 02142, United States
| | - Neil Robinson
- S.H.B. Enterprises Ltd., 55 Station Road, Beaconsfield HP19 1QL, United Kingdom
| | - Jessica A. Rowley
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Angela J. Russell
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3PQ, United Kingdom
| | - Jonathon M. Tinsley
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon OX14 4SB, United Kingdom
| | - Richard Weaver
- XenoGesis Ltd., BioCity Nottingham, Pennyfoot Street, Nottingham NG1 1GF, United Kingdom
| | - Isabel V. L. Wilkinson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Nicky J. Willis
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| | - Francis X. Wilson
- Summit Therapeutics plc, 136a Eastern Avenue, Milton Park, Abingdon OX14 4SB, United Kingdom
| | - Graham M. Wynne
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford OX1 3TA, United Kingdom
| |
Collapse
|
92
|
Fajardo VA, Watson CJF, Bott KN, Moradi F, Maddalena LA, Bellissimo CA, Turner KD, Peters SJ, LeBlanc PJ, MacNeil AJ, Stuart JA, Tupling AR. Neurogranin is expressed in mammalian skeletal muscle and inhibits calcineurin signaling and myoblast fusion. Am J Physiol Cell Physiol 2019; 317:C1025-C1033. [PMID: 31433693 DOI: 10.1152/ajpcell.00345.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Calcineurin is a Ca2+/calmodulin (CaM)-dependent phosphatase that plays a critical role in promoting the slow fiber phenotype and myoblast fusion in skeletal muscle, thereby making calcineurin an attractive cellular target for enhancing fatigue resistance, muscle metabolism, and muscle repair. Neurogranin (Ng) is a CaM-binding protein thought to be expressed solely in brain and neurons, where it inhibits calcineurin signaling by sequestering CaM, thus lowering its cellular availability. Here, we demonstrate for the first time the expression of Ng protein and mRNA in mammalian skeletal muscle. Both protein and mRNA levels are greater in slow-oxidative compared with fast-glycolytic muscles. Coimmunoprecipitation of CaM with Ng in homogenates of C2C12 myotubes, mouse soleus, and human vastus lateralis suggests that these proteins physically interact. To determine whether Ng inhibits calcineurin signaling in muscle, we used Ng siRNA with C2C12 myotubes to reduce Ng protein levels by 60%. As a result of reduced Ng expression, C2C12 myotubes had enhanced CaM-calcineurin binding and calcineurin signaling as indicated by reduced phosphorylation of nuclear factor of activated T cells and increased utrophin mRNA. In addition, calcineurin signaling affects the expression of myogenin and stabilin-2, which are involved in myogenic differentiation and myoblast fusion, respectively. Here, we found that both myogenin and stabilin-2 were significantly elevated by Ng siRNA in C2C12 cells, concomitantly with an increased fusion index. Taken together, these results demonstrate the expression of Ng in mammalian skeletal muscle where it appears to be a novel regulator of calcineurin signaling.
Collapse
Affiliation(s)
- Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Colton J F Watson
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Kirsten N Bott
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
| | - Fereshteh Moradi
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Lucas A Maddalena
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | | | - Kelli D Turner
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Sandra J Peters
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Paul J LeBlanc
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - Adam J MacNeil
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
93
|
Lindsay A, Southern WM, McCourt PM, Larson AA, Hodges JS, Lowe DA, Ervasti JM. Variable cytoplasmic actin expression impacts the sensitivity of different dystrophin-deficient mdx skeletal muscles to eccentric contraction. FEBS J 2019; 286:2562-2576. [PMID: 30942954 PMCID: PMC6613979 DOI: 10.1111/febs.14831] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/24/2019] [Accepted: 04/01/2019] [Indexed: 11/29/2022]
Abstract
Eccentric contractions (ECCs) induce force loss in several skeletal muscles of dystrophin-deficient mice (mdx), with the exception of the soleus (Sol). The eccentric force : isometric force (ECC : ISO), expression level of utrophin, fiber type distribution, and sarcoendoplasmic reticulum calcium ATPase expression are factors that differ between muscles and may contribute to the sensitivity of mdx skeletal muscle to ECC. Here, we confirm that the Sol of mdx mice loses only 13% force compared to 87% in the extensor digitorum longus (EDL) following 10 ECC of isolated muscles. The Sol has a greater proportion of fibers expressing Type I myosin heavy chain (MHC) and expresses 2.3-fold more utrophin compared to the EDL. To examine the effect of ECC : ISO, we show that the mdx Sol is insensitive to ECC at ECC : ISO up to 230 ± 15%. We show that the peroneus longus (PL) muscle presents with similar ECC : ISO compared to the EDL, intermediate force loss (68%) following 10 ECC, and intermediate fiber type distribution and utrophin expression relative to EDL and Sol. The combined absence of utrophin and dystrophin in mdx/utrophin-/- mice rendered the Sol only partially susceptible to ECC and exacerbated force loss in the EDL and PL. Most interestingly, the expression levels of cytoplasmic β- and γ-actins correlate inversely with a given muscle's sensitivity to ECC; EDL < PL < Sol. Our data indicate that fiber type, utrophin, and cytoplasmic actin expression all contribute to the differential sensitivities of mdxEDL, PL, and Sol muscles to ECC.
Collapse
Affiliation(s)
- Angus Lindsay
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - William M. Southern
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Preston M. McCourt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| | - Alexie A. Larson
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, USA
| | - James S. Hodges
- Division of Biostatistics, University of Minnesota, Minneapolis, USA
| | - Dawn A. Lowe
- Division of Rehabilitation Science and Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, USA
| | - James M. Ervasti
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, USA
| |
Collapse
|
94
|
Guiraud S, Edwards B, Babbs A, Squire SE, Berg A, Moir L, Wood MJ, Davies KE. The potential of utrophin and dystrophin combination therapies for Duchenne muscular dystrophy. Hum Mol Genet 2019; 28:2189-2200. [PMID: 30990876 PMCID: PMC6586144 DOI: 10.1093/hmg/ddz049] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/18/2019] [Accepted: 02/26/2019] [Indexed: 01/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal neuromuscular disorder caused by loss of dystrophin. Several therapeutic modalities are currently in clinical trials but none will achieve maximum functional rescue and full disease correction. Therefore, we explored the potential of combining the benefits of dystrophin with increases of utrophin, an autosomal paralogue of dystrophin. Utrophin and dystrophin can be co-expressed and co-localized at the same muscle membrane. Wild-type (wt) levels of dystrophin are not significantly affected by a moderate increase of utrophin whereas higher levels of utrophin reduce wt dystrophin, suggesting a finite number of actin binding sites at the sarcolemma. Thus, utrophin upregulation strategies may be applied to the more mildly affected Becker patients with lower dystrophin levels. Whereas increased dystrophin in wt animals does not offer functional improvement, overexpression of utrophin in wt mice results in a significant supra-functional benefit over wt. These findings highlight an additive benefit of the combined therapy and potential new unique roles of utrophin. Finally, we show a 30% restoration of wt dystrophin levels, using exon-skipping, together with increased utrophin levels restores dystrophic muscle function to wt levels offering greater therapeutic benefit than either single approach alone. Thus, this combination therapy results in additive functional benefit and paves the way for potential future combinations of dystrophin- and utrophin-based strategies.
Collapse
Affiliation(s)
- Simon Guiraud
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Benjamin Edwards
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Arran Babbs
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Sarah E Squire
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Adam Berg
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Lee Moir
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Matthew J Wood
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| | - Kay E Davies
- MDUK Oxford Neuromuscular Centre, Department of Physiology, Anatomy and Genetics, Oxford, UK
| |
Collapse
|
95
|
Randazzo D, Khalique U, Belanto JJ, Kenea A, Talsness DM, Olthoff JT, Tran MD, Zaal KJ, Pak K, Pinal-Fernandez I, Mammen AL, Sackett D, Ervasti JM, Ralston E. Persistent upregulation of the β-tubulin tubb6, linked to muscle regeneration, is a source of microtubule disorganization in dystrophic muscle. Hum Mol Genet 2019; 28:1117-1135. [PMID: 30535187 DOI: 10.1093/hmg/ddy418] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/26/2018] [Accepted: 12/02/2018] [Indexed: 12/20/2022] Open
Abstract
In healthy adult skeletal muscle fibers microtubules form a three-dimensional grid-like network. In the mdx mouse, a model of Duchenne muscular dystrophy (DMD), microtubules are mostly disordered, without periodicity. These microtubule defects have been linked to the mdx mouse pathology. We now report that increased expression of the beta 6 class V β-tubulin (tubb6) contributes to the microtubule changes of mdx muscles. Wild-type muscle fibers overexpressing green fluorescent protein (GFP)-tubb6 (but not GFP-tubb5) have disorganized microtubules whereas mdx muscle fibers depleted of tubb6 (but not of tubb5) normalize their microtubules, suggesting that increasing tubb6 is toxic. However, tubb6 increases spontaneously during differentiation of mouse and human muscle cultures. Furthermore, endogenous tubb6 is not uniformly expressed in mdx muscles but is selectively increased in fiber clusters, which we identify as regenerating. Similarly, mdx-based rescued transgenic mice that retain a higher than expected tubb6 level show focal expression of tubb6 in subsets of fibers. Tubb6 is also upregulated in cardiotoxin-induced mouse muscle regeneration, in human myositis and DMD biopsies, and the tubb6 level correlates with that of embryonic myosin heavy chain, a regeneration marker. In conclusion, modulation of a β-tubulin isotype plays a role in muscle differentiation and regeneration. Increased tubb6 expression and microtubule reorganization are not pathological per se but reflect a return to an earlier developmental stage. However, chronic elevation of tubb6, as occurs in the mdx mouse, may contribute to the repeated cycles of regeneration and to the pathology of the disease.
Collapse
Affiliation(s)
- Davide Randazzo
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Umara Khalique
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Joseph J Belanto
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Aster Kenea
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dana M Talsness
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - John T Olthoff
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Michelle D Tran
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kristien J Zaal
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine Pak
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA
| | - Iago Pinal-Fernandez
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA.,Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Andrew L Mammen
- Laboratory of Muscle Stem Cells and Gene Regulation, Muscle Disease Unit, NIAMS, NIH, Bethesda, MD, USA.,Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan Sackett
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), NIH, Bethesda, MD, USA
| | - James M Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, and Program in Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota-Twin Cities, Minneapolis, MN, USA
| | - Evelyn Ralston
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
96
|
Soluble Heparin Binding Epidermal Growth Factor-Like Growth Factor Is a Regulator of GALGT2 Expression and GALGT2-Dependent Muscle and Neuromuscular Phenotypes. Mol Cell Biol 2019; 39:MCB.00140-19. [PMID: 31036568 DOI: 10.1128/mcb.00140-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 04/26/2019] [Indexed: 02/08/2023] Open
Abstract
GALGT2 (also B4GALNT2) encodes a glycosyltransferase that is normally confined to the neuromuscular and myotendinous junction in adult skeletal muscle. GALGT2 overexpression in muscle can inhibit muscular dystrophy in mouse models of the disease by inducing the overexpression of surrogate muscle proteins, including utrophin, agrin, laminins, and integrins. Despite its well-documented biological properties, little is known about the endogenous regulation of muscle GALGT2 expression. Here, we demonstrate that epidermal growth factor receptor (EGFR) ligands can activate the human GALGT2 promoter. Overexpression of one such ligand, soluble heparin-binding EGF-like growth factor (sHB-EGF), also stimulated mouse muscle Galgt2 gene expression and expression of GALGT2-inducible surrogate muscle genes. Deletion analysis of the GALGT2 promoter identified a 45-bp region containing a TFAP4-binding site that was required for sHB-EGF activation. sHB-EGF increased TFAP4 binding to this site in muscle cells and increased endogenous Tfap4 gene expression. sHB-EGF also increased muscle EGFR protein expression and activated EGFR-Akt signaling. sHB-EGF expression was concentrated at the neuromuscular junction, and Hbegf deletion reduced Galgt2-dependent synaptic glycosylation. Hbegf deletion also mimicked Galgt2-dependent neuromuscular and muscular dystrophy phenotypes. These data demonstrate that sHB-EGF is an endogenous regulator of muscle Galgt2 gene expression and can mimic Galgt2-dependent muscle phenotypes.
Collapse
|
97
|
Guiraud S, Edwards B, Squire SE, Moir L, Berg A, Babbs A, Ramadan N, Wood MJ, Davies KE. Embryonic myosin is a regeneration marker to monitor utrophin-based therapies for DMD. Hum Mol Genet 2019; 28:307-319. [PMID: 30304405 PMCID: PMC6322073 DOI: 10.1093/hmg/ddy353] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/28/2018] [Indexed: 11/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked muscle-wasting disease caused by lack of the cytoskeletal protein dystrophin. Constitutive utrophin expression, a structural and functional paralogue of dystrophin, can successfully prevent the dystrophic pathology in the dystrophin-deficient mdx mouse model. In dystrophic muscles, utrophin is increased as part of the repair process and localized at the sarcolemma of regenerating myofibers. The presence of developmental myosin such as embryonic myosin (MyHC-emb) and neonatal represents a useful marker of muscle regeneration and a meaningful indicator of muscle damage, which correlates with the clinical severity of milder Becker muscular dystrophy and DMD patients. In the present study, we demonstrate that MyHC-emb is a robust marker of regeneration at different ages and in different skeletal muscles. We also evaluate the correlation between utrophin, dystrophin and MyHC-emb in wild-type (wt) and regenerating dystrophic muscles. Restoration of dystrophin significantly reduced MyHC-emb levels. Similarly, overexpression of utrophin in the transgenic mdx-Fiona mice reduced the number of MyHC-emb positive fibers to wt level, prevented the regenerative process and rescued the muscle function. In contrast, the absence of utrophin in the dystrophin-deficient double-knockout mice resulted in a higher MyHC-emb content and in a more severe dystrophic pathophysiology than in mdx mice. These data illustrate the importance of monitoring utrophin and MyHC-emb levels in the preclinical evaluation of therapies and provide translational support for the use of developmental myosin as a disease biomarker in DMD clinical trials.
Collapse
Affiliation(s)
- Simon Guiraud
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Benjamin Edwards
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Sarah E Squire
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Lee Moir
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Adam Berg
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Arran Babbs
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Nesrine Ramadan
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Matthew J Wood
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Kay E Davies
- Department of Physiology, Anatomy and Genetics, MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
98
|
Cai A, Kong X. Development of CRISPR-Mediated Systems in the Study of Duchenne Muscular Dystrophy. Hum Gene Ther Methods 2019; 30:71-80. [PMID: 31062609 DOI: 10.1089/hgtb.2018.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe type of X-linked recessive degenerative muscle disease caused by mutations in the dystrophin (DMD) gene on the X chromosome. The DMD gene is complex, consisting of 79 exons, and mutations cause changes in the DMD mRNA so that the reading frame is altered, and the muscle-specific isoform of the dystrophin protein is either absent or truncated with variable residual function. The emerging CRISPR-Cas9-mediated genome editing technique is being developed as a potential therapeutic approach to treat DMD because it can permanently replace the mutated dystrophin gene with the normal gene. Prenatal DNA testing can inform whether the female fetus is a carrier of DMD, and the male fetus has inherited a mutation from his mother (50% chance of both). This article summarizes the present status of current and future treatments for DMD.
Collapse
Affiliation(s)
- Aojie Cai
- Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Xiangdong Kong
- Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
99
|
Fajardo VA, Chambers PJ, Juracic ES, Rietze BA, Gamu D, Bellissimo C, Kwon F, Quadrilatero J, Russell Tupling A. Sarcolipin deletion in mdx mice impairs calcineurin signalling and worsens dystrophic pathology. Hum Mol Genet 2019; 27:4094-4102. [PMID: 30137316 DOI: 10.1093/hmg/ddy302] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most severe form of muscular dystrophy affecting 1 in 3500 live male births. Although there is no cure for DMD, therapeutic strategies aimed at enhancing calcineurin signalling and promoting the slow fibre phenotype have shown promise in mdx mice, which is the classical mouse model for DMD. Sarcolipin (SLN) is a small protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase pump and its expression is highly upregulated in dystrophic skeletal muscle. We have recently shown that SLN in skeletal muscle amplifies calcineurin signalling thereby increasing myofibre size and the slow fibre phenotype. Therefore, in the present study we sought to determine the physiological impact of genetic Sln deletion in mdx mice, particularly on calcineurin signalling, fibre-type distribution and size and dystrophic pathology. We generated an mdx/Sln-null (mdx/SlnKO) mouse colony and hypothesized that the soleus and diaphragm muscles from these mice would display blunted calcineurin signalling, smaller myofibre sizes, an increased proportion of fast fibres and worsened dystrophic pathology compared with mdx mice. Our results show that calcineurin signalling was impaired in mdx/SlnKO mice as indicated by reductions in utrophin, stabilin-2 and calcineurin expression. In addition, mdx/SlnKO muscles contained smaller myofibres, exhibited a slow-to-fast fibre-type switch that corresponded with reduced expression of mitochondrial proteins and displayed a worsened dystrophic pathology compared with mdx muscles. Altogether, our findings demonstrate a critical role for SLN upregulation in dystrophic muscles and suggest that SLN can be viewed as a potential therapeutic target.
Collapse
Affiliation(s)
- Val A Fajardo
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Paige J Chambers
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Emma S Juracic
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Bradley A Rietze
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Daniel Gamu
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | | | - Frenk Kwon
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| |
Collapse
|
100
|
Parvatiyar MS, Brownstein AJ, Kanashiro-Takeuchi RM, Collado JR, Dieseldorff Jones KM, Gopal J, Hammond KG, Marshall JL, Ferrel A, Beedle AM, Chamberlain JS, Renato Pinto J, Crosbie RH. Stabilization of the cardiac sarcolemma by sarcospan rescues DMD-associated cardiomyopathy. JCI Insight 2019; 5:123855. [PMID: 31039133 PMCID: PMC6629091 DOI: 10.1172/jci.insight.123855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 04/23/2019] [Indexed: 02/02/2023] Open
Abstract
In the current preclinical study, we demonstrate the therapeutic potential of sarcospan (SSPN) overexpression to alleviate cardiomyopathy associated with Duchenne muscular dystrophy (DMD) utilizing dystrophin-deficient mdx mice with utrophin haploinsufficiency that more accurately represent the severe disease course of human DMD. SSPN interacts with dystrophin, the DMD disease gene product, and its autosomal paralog utrophin, which is upregulated in DMD as a partial compensatory mechanism. SSPN transgenic mice have enhanced abundance of fully glycosylated α-dystroglycan, which may further protect dystrophin-deficient cardiac membranes. Baseline echocardiography reveals SSPN improves systolic function and hypertrophic indices in mdx and mdx:utr-heterozygous mice. Assessment of SSPN transgenic mdx mice by hemodynamic pressure-volume methods highlights enhanced systolic performance compared to mdx controls. SSPN restores cardiac sarcolemma stability, the primary defect in DMD disease, reduces fibrotic response and improves contractile function. We demonstrate that SSPN ameliorates more advanced cardiac disease in the context of diminished sarcolemma expression of utrophin and β1D integrin that mitigate disease severity and partially restores responsiveness to β-adrenergic stimulation. Overall, our current and previous findings suggest SSPN overexpression in DMD mouse models positively impacts skeletal, pulmonary and cardiac performance by addressing the stability of proteins at the sarcolemma that protect the heart from injury, supporting SSPN and membrane stabilization as a therapeutic target for DMD.
Collapse
Affiliation(s)
- Michelle S. Parvatiyar
- Department of Integrative Biology & Physiology and
- Center for Duchenne Muscular Dystrophy, UCLA, Los Angeles, California, USA
| | - Alexandra J. Brownstein
- Department of Integrative Biology & Physiology and
- Center for Duchenne Muscular Dystrophy, UCLA, Los Angeles, California, USA
| | - Rosemeire M. Kanashiro-Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami, Florida, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | | | - Jay Gopal
- Department of Integrative Biology & Physiology and
| | - Katherine G. Hammond
- Department of Integrative Biology & Physiology and
- Center for Duchenne Muscular Dystrophy, UCLA, Los Angeles, California, USA
| | - Jamie L. Marshall
- Department of Integrative Biology & Physiology and
- Center for Duchenne Muscular Dystrophy, UCLA, Los Angeles, California, USA
| | - Abel Ferrel
- Department of Integrative Biology & Physiology and
| | - Aaron M. Beedle
- Department of Pharmaceutical Sciences, Binghamton University State University of New York, Binghamton, New York, USA
| | | | - Jose Renato Pinto
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Rachelle H. Crosbie
- Department of Integrative Biology & Physiology and
- Center for Duchenne Muscular Dystrophy, UCLA, Los Angeles, California, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Molecular Biology Institute, UCLA, Los Angeles, California, USA
| |
Collapse
|