101
|
Park S, Maloney B, Caggana M, Tavakoli NP. Creatine kinase-MM concentration in dried blood spots from newborns and implications for newborn screening for Duchenne muscular dystrophy. Muscle Nerve 2022; 65:652-658. [PMID: 35307847 PMCID: PMC9322420 DOI: 10.1002/mus.27533] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
Introduction/Aims Creatine kinase‐MM (CK‐MM) is a marker of skeletal muscle damage. Detection of elevated levels of CK‐MM in newborns can enable an early suspicion of the diagnosis of Duchenne muscular dystrophy (DMD) before symptom onset. Our aim was to investigate CK‐MM levels in DMD‐affected and unaffected newborns using an immunoassay that measures CK‐MM concentration in dried blood spots collected for routine newborn screening. Methods To validate the assay in our laboratory, CK‐MM measurements and newborn demographic information were collected for 8584 de‐identified specimens and 15 confirmed DMD patients. After analyzing validation data, CK‐MM normal ranges were determined based on age of newborn at specimen collection. Subsequently, the assay was used to measure CK‐MM concentration in 26 135 newborns as part of a consented pilot study to screen for DMD in New York State. Mean and median levels of CK‐MM based on age of collection, in addition to the 2.5th, 50th, 97.5th, and 99.5th percentiles, were recalculated using the validation and screening data sets. Results Median CK‐MM within 1 hour of birth was 109 ng/mL, rose to a high of 499 ng/mL at 25 hours of age, and then declined to 200 ng/mL at 2 days of life. The median continued to decline more slowly and then stabilized at approximately 40 ng/mL at 1 week of life. Discussion Because of the marked variability and elevated CK‐MM levels observed within the first days of life, it is important to set multiple CK‐MM age‐related cut‐offs when screening for DMD in newborns.
Collapse
Affiliation(s)
- Sunju Park
- Division of Genetics, Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, New York, USA
| | - Breanne Maloney
- Division of Genetics, Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, New York, USA
| | - Michele Caggana
- Division of Genetics, Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, New York, USA
| | - Norma P Tavakoli
- Division of Genetics, Wadsworth Center, New York State Department of Health, David Axelrod Institute, Albany, New York, USA
| |
Collapse
|
102
|
Mhandire DZ, Burns DP, Roger AL, O'Halloran KD, ElMallah MK. Breathing in Duchenne muscular dystrophy: Translation to therapy. J Physiol 2022; 600:3465-3482. [PMID: 35620971 PMCID: PMC9357048 DOI: 10.1113/jp281671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/17/2022] [Indexed: 11/08/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a deficiency in dystrophin - a structural protein which stabilizes muscle during contraction. Dystrophin deficiency adversely affects the respiratory system leading to sleep-disordered breathing, hypoventilation, and weakness of the expiratory and inspiratory musculature, which culminate in severe respiratory dysfunction. Muscle degeneration associated respiratory impairment in neuromuscular disease is a result of disruptions at multiple sites of the respiratory control network, including sensory and motor pathways. As a result of this pathology, respiratory failure is a leading cause of premature death in DMD patients. Currently available treatments for DMD respiratory insufficiency attenuate respiratory symptoms without completely reversing the underlying pathophysiology. This underscores the need to develop curative therapies to improve quality of life and longevity of DMD patients. This review summarises research findings on the pathophysiology of respiratory insufficiencies in DMD disease in humans and animal models, the clinical interventions available to ameliorate symptoms, and gene-based therapeutic strategies uncovered by preclinical animal studies. Abstract figure legend: Summary of the therapeutic strategies for respiratory insufficiency in DMD (Duchenne muscular dystrophy). Treatment options currently in clinical use only attenuate respiratory symptoms without reversing the underlying pathology of DMD-associated respiratory insufficiencies. Ongoing preclinical and clinical research is aimed at developing curative therapies that both improve quality of life and longevity of DMD patients. AAV - adeno-associated virus, PPMO - Peptide-conjugated phosphorodiamidate morpholino oligomer This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Doreen Z Mhandire
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Angela L Roger
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Mai K ElMallah
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Duke University Medical Center Box 2644, Durham, North Carolina, 27710, USA
| |
Collapse
|
103
|
Clemens PR, Rao VK, Connolly AM, Harper AD, Mah JK, McDonald CM, Smith EC, Zaidman CM, Nakagawa T, Hoffman EP. Long-Term Functional Efficacy and Safety of Viltolarsen in Patients with Duchenne Muscular Dystrophy. J Neuromuscul Dis 2022; 9:493-501. [PMID: 35634851 PMCID: PMC9398057 DOI: 10.3233/jnd-220811] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Duchenne muscular dystrophy (DMD) is a rare, genetic disease caused by mutations in the DMD gene resulting in an absence of functional dystrophin protein. Viltolarsen, an exon 53 skipping therapy, has been shown to increase endogenous dystrophin levels. Herein, long-term (>2 years) functional outcomes in viltolarsen treated patients were compared to a matched historical control group. Objective: To evaluate long-term efficacy and safety of the anti-sense oligonucleotide viltolarsen in the treatment of patients with DMD amenable to exon 53 skipping therapy. Methods: This trial (NCT03167255) is the extension of a previously published 24-week trial in North America (NCT02740972) that examined dystrophin levels, timed function tests compared to a matched historical control group (Cooperative International Neuromuscular Research Group Duchenne Natural History Study, CINRG DNHS), and safety in boys 4 to < 10 years (N = 16) with DMD amenable to exon 53 skipping who were treated with viltolarsen. Both groups were treated with glucocorticoids. All 16 participants elected to enroll in this long-term trial (up to 192 weeks) to continue evaluation of motor function and safety. Results: Time to stand from supine and time to run/walk 10 meters showed stabilization from baseline through week 109 for viltolarsen-treated participants whereas the historical control group showed decline (statistically significant differences for multiple timepoints). Safety was similar to that observed in the previous 24-week trial, which was predominantly mild. There have been no treatment-related serious adverse events and no discontinuations. Conclusions: Based on these results at over 2 years, viltolarsen can be a new treatment option for patients with DMD amenable to exon 53 skipping.
Collapse
Affiliation(s)
- Paula R. Clemens
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Veterans Affairs Medical Center, Pittsburgh, Pennsylvania
| | - Vamshi K. Rao
- Division of Neurology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois
| | - Anne M. Connolly
- Division of Neurology, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, Ohio
| | - Amy D. Harper
- Children’s Hospital of Richmond at Virginia Commonwealth University, Richmond, Virginia
| | - Jean K. Mah
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Craig M. McDonald
- Department of Physical Medicine and Rehabilitation, Department of Pediatrics, UC Davis Health, University of California, Davis, Sacramento, California
| | | | - Craig M. Zaidman
- Department of Neurology, Washington University at St Louis, St Louis, Missouri
| | | | - Eric P. Hoffman
- Department of Pharmaceutical Sciences, State University of New York at Binghamton, Binghamton, New York
| | | |
Collapse
|
104
|
Long-Term Protective Effect of Human Dystrophin Expressing Chimeric (DEC) Cell Therapy on Amelioration of Function of Cardiac, Respiratory and Skeletal Muscles in Duchenne Muscular Dystrophy. Stem Cell Rev Rep 2022; 18:2872-2892. [PMID: 35590083 PMCID: PMC9622520 DOI: 10.1007/s12015-022-10384-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2022] [Indexed: 12/12/2022]
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal disease caused by mutations in dystrophin encoding gene, causing progressive degeneration of cardiac, respiratory, and skeletal muscles leading to premature death due to cardiac and respiratory failure. Currently, there is no cure for DMD. Therefore, novel therapeutic approaches are needed for DMD patients. We have previously reported functional improvements which correlated with increased dystrophin expression following administration of dystrophin expressing chimeric (DEC) cells of myoblast origin to the mdx mouse models of DMD. In the current study, we confirmed dose-dependent protective effect of human DEC therapy created from myoblasts of normal and DMD-affected donors, on restoration of dystrophin expression and amelioration of cardiac, respiratory, and skeletal muscle function at 180 days after systemic-intraosseous DEC administration to mdx/scid mouse model of DMD. Functional improvements included maintenance of ejection fraction and fractional shortening levels on echocardiography, reduced enhanced pause and expiration time on plethysmography and improved grip strength and maximum stretch induced contraction of skeletal muscles. Improved function was associated with amelioration of mdx muscle pathology revealed by reduced muscle fibrosis, reduced inflammation and improved muscle morphology confirmed by reduced number of centrally nucleated fibers and normalization of muscle fiber diameters. Our findings confirm the long-term systemic effect of DEC therapy in the most severely affected by DMD organs including heart, diaphragm, and long skeletal muscles. These encouraging preclinical data introduces human DEC as a novel therapeutic modality of Advanced Therapy Medicinal Product (ATMP) with the potential to improve or halt the progression of DMD and enhance quality of life of DMD patients.
Collapse
|
105
|
Del Rio-Pertuz G, Morataya C, Parmar K, Dubay S, Argueta-Sosa E. Dilated cardiomyopathy as the initial presentation of Becker muscular dystrophy: a systematic review of published cases. Orphanet J Rare Dis 2022; 17:194. [PMID: 35549971 PMCID: PMC9097097 DOI: 10.1186/s13023-022-02346-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/26/2022] [Indexed: 11/10/2022] Open
Abstract
There are scarce publications regarding the presentation and outcome of Becker muscular dystrophy in adulthood when idiopathic dilated cardiomyopathy is the initial disease manifestation. We performed a systematic review using Medline, Embase, Cochrane, and Scopus to identify cases of adults with idiopathic dilated cardiomyopathy who were subsequently diagnosed with Becker muscular dystrophy from inception through August 2020. Six cases were found. We identified young males (Median age: 26 years) with Becker muscular dystrophy who first presented with dilated cardiomyopathy. Most patients initially presented with congestive heart failure symptoms (5/6, 83%), and had a median left ventricular ejection fraction of 23%. One case did have calf pseudohypertrophy. Musculoskeletal symptoms later appeared one to six years after the initial dilated cardiomyopathy presentation. Heart transplantation was the most common management strategy (4/6, 67%). A left ventricular assist device was used in one case as a bridge to heart transplant. Dilated cardiomyopathy can be the initial presentation of Becker muscular dystrophy in the third to fourth decades of life in adult patients, and musculoskeletal symptoms can be subclinical.
Collapse
Affiliation(s)
- Gaspar Del Rio-Pertuz
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430, USA.
| | - Cristina Morataya
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430, USA
| | - Kanak Parmar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430, USA
| | - Sarah Dubay
- Department of Clinical and Diagnostic Sciences, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erwin Argueta-Sosa
- Division of Cardiology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
106
|
Saifullah, Motohashi N, Tsukahara T, Aoki Y. Development of Therapeutic RNA Manipulation for Muscular Dystrophy. Front Genome Ed 2022; 4:863651. [PMID: 35620642 PMCID: PMC9127466 DOI: 10.3389/fgeed.2022.863651] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Approval of therapeutic RNA molecules, including RNA vaccines, has paved the way for next-generation treatment strategies for various diseases. Oligonucleotide-based therapeutics hold particular promise for treating incurable muscular dystrophies, including Duchenne muscular dystrophy (DMD). DMD is a severe monogenic disease triggered by deletions, duplications, or point mutations in the DMD gene, which encodes a membrane-linked cytoskeletal protein to protect muscle fibers from contraction-induced injury. Patients with DMD inevitably succumb to muscle degeneration and atrophy early in life, leading to premature death from cardiac and respiratory failure. Thus far, the disease has thwarted all curative strategies. Transcriptomic manipulation, employing exon skipping using antisense oligonucleotides (ASO), has made significant progress in the search for DMD therapeutics. Several exon-skipping drugs employing RNA manipulation technology have been approved by regulatory agencies and have shown promise in clinical trials. This review summarizes recent scientific and clinical progress of ASO and other novel RNA manipulations, including RNA-based editing using MS2 coat protein-conjugated adenosine deaminase acting on the RNA (MCP-ADAR) system illustrating the efficacy and limitations of therapies to restore dystrophin. Perhaps lessons from this review will encourage the application of RNA-editing therapy to other neuromuscular disorders.
Collapse
Affiliation(s)
- Saifullah
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Toshifumi Tsukahara
- Area of Bioscience and Biotechnology, School of Materials Science, Japan Advanced Institute of Science and Technology (JAIST), Ishikawa, Japan
- Division of Transdisciplinary Science, Japan Advanced Institute of Science and Technology (JAIST), Ishikawa, Japan
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| |
Collapse
|
107
|
Colapicchioni V, Millozzi F, Parolini O, Palacios D. Nanomedicine, a valuable tool for skeletal muscle disorders: Challenges, promises, and limitations. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1777. [PMID: 35092179 PMCID: PMC9285803 DOI: 10.1002/wnan.1777] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 12/15/2022]
Abstract
Muscular dystrophies are a group of rare genetic disorders characterized by progressive muscle weakness, which, in the most severe forms, leads to the patient's death due to cardiorespiratory problems. There is still no cure available for these diseases and significant effort is being placed into developing new strategies to either correct the genetic defect or to compensate muscle loss by stimulating skeletal muscle regeneration. However, the vast anatomical extension of the target tissue poses great challenges to these goals, highlighting the need for complementary strategies. Nanomedicine is an actively evolving field that merges nanotechnologies with biomedical and pharmaceutical sciences. It holds great potential in regenerative medicine, both in supporting tissue engineering and regeneration, and in optimizing drug and oligonucleotide delivery and gene therapy strategies. In this review, we will summarize the state‐of‐the‐art in the field of nanomedicine applied to skeletal muscle regeneration. We will discuss the recent work toward the development of nanopatterned scaffolds for tissue engineering, the efforts in the synthesis of organic and inorganic nanoparticles for gene therapy and drug delivery applications, as well as their use as immune modulators. Although nanomedicine holds great promise for muscle and other degenerative diseases, many challenges still need to be systematically addressed to assure a smooth transition from the bench to the bedside. This article is categorized under:Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement
Collapse
Affiliation(s)
- Valentina Colapicchioni
- Italian National Research Council, Institute for Atmospheric Pollution Research (CNR-IIA), Rome, Italy.,Mhetra LLC, Miami, Florida, USA
| | - Francesco Millozzi
- Histology and Embryology Unit, DAHFMO, Sapienza University, Rome, Italy.,IRCCS Santa Lucia Foundation, Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy.,IRCCS Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
108
|
Zheng F, Chen Z, Li J, Wu R, Zhang B, Nie G, Xie Z, Zhang H. A Highly Sensitive CRISPR-Empowered Surface Plasmon Resonance Sensor for Diagnosis of Inherited Diseases with Femtomolar-Level Real-Time Quantification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105231. [PMID: 35343100 PMCID: PMC9108660 DOI: 10.1002/advs.202105231] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/21/2022] [Indexed: 05/25/2023]
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR) molecular system has emerged as a promising technology for the detection of nucleic acids. Herein, the development of a surface plasmon resonance (SPR) sensor that is functionalized with a layer of locally grown graphdiyne film, achieving excellent sensing performance when coupled with catalytically deactivated CRISPR-associated protein 9 (dCas9), is reported. dCas9 protein is immobilized on the sensor surface and complexed with a specific single-guide RNA, enabling the amplification-free detection of target sequences within genomic DNA. The sensor, termed CRISPR-SPR-Chip, is used to successfully analyze recombinant plasmids with only three-base mutations with a limit of detection as low as 1.3 fM. Real-time monitoring CRISPR-SPR-Chip is used to analyze clinical samples of patients with Duchenne muscular dystrophy with two exon deletions, which are detected without any pre-amplification step, yielding significantly positive results within 5 min. The ability of this novel CRISPR-empowered SPR (CRISPR-eSPR) sensing platform to rapidly, precisely, sensitively, and specifically detect a target gene sequence provides a new on-chip optic approach for clinical gene analysis.
Collapse
Affiliation(s)
- Fei Zheng
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
| | - Zhi Chen
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
| | - Jingfeng Li
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
- Shenzhen International Institute for Biomedical ResearchShenzhen518110China
| | - Rui Wu
- Laboratory of Robotics and SystemHarbin Institute of TechnologyHarbin150001P. R. China
| | - Bin Zhang
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
| | - Guohui Nie
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
| | - Zhongjian Xie
- Institute of PediatricsShenzhen Children's HospitalShenzhenGuangdong518038P. R. China
- Shenzhen International Institute for Biomedical ResearchShenzhenGuangdong518116P. R. China
| | - Han Zhang
- Shenzhen Engineering Laboratory of phosphorene and OptoelectronicsInternational Collaborative Laboratory of 2D Materials for Optoelectronics Science and Technology of Ministry of EducationShenzhen Institute of Translational MedicineDepartment of OtolaryngologyShenzhen Second People's Hospitalthe First Affiliated HospitalInstitute of Microscale OptoelectronicsShenzhen UniversityShenzhen518060P.R. China
| |
Collapse
|
109
|
Guglieri M, Bushby K, McDermott MP, Hart KA, Tawil R, Martens WB, Herr BE, McColl E, Speed C, Wilkinson J, Kirschner J, King WM, Eagle M, Brown MW, Willis T, Griggs RC. Effect of Different Corticosteroid Dosing Regimens on Clinical Outcomes in Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. JAMA 2022; 327:1456-1468. [PMID: 35381069 PMCID: PMC8984930 DOI: 10.1001/jama.2022.4315] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE Corticosteroids improve strength and function in boys with Duchenne muscular dystrophy. However, there is uncertainty regarding the optimum regimen and dosage. OBJECTIVE To compare efficacy and adverse effects of the 3 most frequently prescribed corticosteroid regimens in boys with Duchenne muscular dystrophy. DESIGN, SETTING, AND PARTICIPANTS Double-blind, parallel-group randomized clinical trial including 196 boys aged 4 to 7 years with Duchenne muscular dystrophy who had not previously been treated with corticosteroids; enrollment occurred between January 30, 2013, and September 17, 2016, at 32 clinic sites in 5 countries. The boys were assessed for 3 years (last participant visit on October 16, 2019). INTERVENTIONS Participants were randomized to daily prednisone (0.75 mg/kg) (n = 65), daily deflazacort (0.90 mg/kg) (n = 65), or intermittent prednisone (0.75 mg/kg for 10 days on and then 10 days off) (n = 66). MAIN OUTCOMES AND MEASURES The global primary outcome comprised 3 end points: rise from the floor velocity (in rise/seconds), forced vital capacity (in liters), and participant or parent global satisfaction with treatment measured by the Treatment Satisfaction Questionnaire for Medication (TSQM; score range, 0 to 100), each averaged across all study visits after baseline. Pairwise group comparisons used a Bonferroni-adjusted significance level of .017. RESULTS Among the 196 boys randomized (mean age, 5.8 years [SD, 1.0 years]), 164 (84%) completed the trial. Both daily prednisone and daily deflazacort were more effective than intermittent prednisone for the primary outcome (P < .001 for daily prednisone vs intermittent prednisone using a global test; P = .017 for daily deflazacort vs intermittent prednisone using a global test) and the daily regimens did not differ significantly (P = .38 for daily prednisone vs daily deflazacort using a global test). The between-group differences were principally attributable to rise from the floor velocity (0.06 rise/s [98.3% CI, 0.03 to 0.08 rise/s] for daily prednisone vs intermittent prednisone [P = .003]; 0.06 rise/s [98.3% CI, 0.03 to 0.09 rise/s] for daily deflazacort vs intermittent prednisone [P = .017]; and -0.004 rise/s [98.3% CI, -0.03 to 0.02 rise/s] for daily prednisone vs daily deflazacort [P = .75]). The pairwise comparisons for forced vital capacity and TSQM global satisfaction subscale score were not statistically significant. The most common adverse events were abnormal behavior (22 [34%] in the daily prednisone group, 25 [38%] in the daily deflazacort group, and 24 [36%] in the intermittent prednisone group), upper respiratory tract infection (24 [37%], 19 [29%], and 24 [36%], respectively), and vomiting (19 [29%], 17 [26%], and 15 [23%]). CONCLUSIONS AND RELEVANCE Among patients with Duchenne muscular dystrophy, treatment with daily prednisone or daily deflazacort, compared with intermittent prednisone alternating 10 days on and 10 days off, resulted in significant improvement over 3 years in a composite outcome comprising measures of motor function, pulmonary function, and satisfaction with treatment; there was no significant difference between the 2 daily corticosteroid regimens. The findings support the use of a daily corticosteroid regimen over the intermittent prednisone regimen tested in this study as initial treatment for boys with Duchenne muscular dystrophy. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01603407.
Collapse
Affiliation(s)
- Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Kate Bushby
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Michael P. McDermott
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York
| | - Kimberly A. Hart
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - William B. Martens
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Barbara E. Herr
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | | | - Chris Speed
- Newcastle University, Newcastle upon Tyne, England
- NIHR Clinical Research Network North East and North Cumbria, Newcastle upon Tyne, England
| | | | - Janbernd Kirschner
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, University Hospital Freiburg, Freiburg, Germany
- Department of Neuropediatrics, University Hospital Bonn, Bonn, Germany
| | | | - Michelle Eagle
- John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England
| | - Mary W. Brown
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | - Tracey Willis
- Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, England
| | - Robert C. Griggs
- Department of Neurology, University of Rochester Medical Center, Rochester, New York
| | | |
Collapse
|
110
|
Functional analysis of variants in DMD exon/intron 10 predicted to affect splicing. J Hum Genet 2022; 67:495-501. [DOI: 10.1038/s10038-022-01035-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 11/08/2022]
|
111
|
Antisense Oligonucleotides Conjugated with Lipophilic Compounds: Synthesis and In Vitro Evaluation of Exon Skipping in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23084270. [PMID: 35457088 PMCID: PMC9032562 DOI: 10.3390/ijms23084270] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/04/2023] Open
Abstract
Our groups previously reported that conjugation at 3′-end with ursodeoxycholic acid (UDCA) significantly enhanced in vitro exon skipping properties of ASO 51 oligonucleotide targeting the human DMD exon 51. In this study, we designed a series of lipophilic conjugates of ASO 51, to explore the influence of the lipophilic moiety on exon skipping efficiency. To this end, three bile acids and two fatty acids have been derivatized and/or modified and conjugated to ASO 51 by automatized solid phase synthesis. We measured the melting temperature (Tm) of lipophilic conjugates to evaluate their ability to form a stable duplex with the target RNA. The exon skipping efficiency has been evaluated in myogenic cell lines first in presence of a transfection agent, then in gymnotic conditions on a selection of conjugated ASO 51. In the case of 5′-UDC-ASO 51, we also evaluated the influence of PS content on exon skipping efficiency; we found that it performed better exon skipping with full PS linkages. The more efficient compounds in terms of exon skipping were found to be 5′-UDC- and 5′,3′-bis-UDC-ASO 51.
Collapse
|
112
|
Kotov AA, Bazylev SS, Adashev VE, Shatskikh AS, Olenina LV. Drosophila as a Model System for Studying of the Evolution and Functional Specialization of the Y Chromosome. Int J Mol Sci 2022; 23:4184. [PMID: 35457001 PMCID: PMC9031259 DOI: 10.3390/ijms23084184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023] Open
Abstract
The Y chromosome is one of the sex chromosomes found in males of animals of different taxa, including insects and mammals. Among all chromosomes, the Y chromosome is characterized by a unique chromatin landscape undergoing dynamic evolutionary change. Being entirely heterochromatic, the Y chromosome as a rule preserves few functional genes, but is enriched in tandem repeats and transposons. Due to difficulties in the assembly of the highly repetitive Y chromosome sequence, deep analyses of Y chromosome evolution, structure, and functions are limited to a few species, one of them being Drosophila melanogaster. Despite Y chromosomes exhibiting high structural divergence between even closely related species, Y-linked genes have evolved convergently and are mainly associated with spermatogenesis-related activities. This indicates that male-specific selection is a dominant force shaping evolution of Y chromosomes across species. This review presents our analysis of current knowledge concerning Y chromosome functions, focusing on recent findings in Drosophila. Here we dissect the experimental and bioinformatics data about the Y chromosome accumulated to date in Drosophila species, providing comparative analysis with mammals, and discussing the relevance of our analysis to a wide range of eukaryotic organisms, including humans.
Collapse
Affiliation(s)
| | | | | | | | - Ludmila V. Olenina
- Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (A.A.K.); (S.S.B.); (V.E.A.); (A.S.S.)
| |
Collapse
|
113
|
Abdulhady H, Sakr HM, Elsayed NS, El-Sobky TA, Fahmy N, Saadawy AM, Elsedfy H. Ambulatory Duchenne muscular dystrophy children: cross-sectional correlation between function, quantitative muscle ultrasound and MRI. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2022; 41:1-14. [PMID: 35465338 PMCID: PMC9004336 DOI: 10.36185/2532-1900-063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/11/2022] [Indexed: 01/24/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive genetic muscle disease. Quantitative muscle ultrasound (US), muscle MRI, and functional tools are important to delineate characteristics of muscle involvement. We aimed to establish correlations between clinical/functional and above-named imaging tools respecting their diagnostic and prognostic role in DMD children. A cross-sectional retrospective study of 27 steroid-naive, ambulant male children/adolescents with genetically-confirmed DMD (mean age, 8.8 ± 3.3 years). Functional performance was assessed using motor function measure (MFM) which assess standing/transfer (D1), proximal (D2) and distal (D3) motor function, and six-minute walk test (6MWT). Imaging evaluation included quantitative muscle MRI which measured muscle fat content in a specific location of right rectus femoris by mDixon sequence. Quantitative muscle US measured right rectus femoris muscle brightness in standardized US image as an indicator of muscle fat content. We found a highly significant positive correlation between the mean MFM total score and 6MWT (R = 0.537, p = 0.007), and a highly significant negative correlation between fat content by muscle US and MFM total score (R = -0.603, p = 0.006) and its D1 subscore (R =-0.712, p = 0.001), and a significant negative correlation between fat content by US and 6MWT (R = -0.529, p = 0.02), and a significant positive correlation between muscle fat content by mDixon MRI and patient's age (R = 0.617, p = 0.01). Quantitative muscle US correlates significantly with clinical/functional assessment tools as MFM and 6MWT, and augments their role in disease-tracking of DMD. Quantitative muscle US has the potential to act as a substitute to functional assessment tools.
Collapse
Affiliation(s)
- Hala Abdulhady
- Department of Physical Medicine, Rheumatology and Rehabilitation, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hossam M. Sakr
- Department of Diagnostic and Interventional Radiology and Molecular Imaging, Faculty of Medicine, Ain Shams University, Cairo, Egypt,Correspondence Hossam M. Sakr Department of Diagnostic and Interventional Radiology and Molecular Imaging Faculty of Medicine, Ain Shams University, Abbassia square, 11381 Cairo, Egypt. E-mail:
| | - Nermine S. Elsayed
- Department of Medical Genetics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Tamer A. El-Sobky
- Division of Pediatric Orthopedics, Department of Orthopedic Surgery, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nagia Fahmy
- Neuromuscular Unit, Department of Neuropsychiatry, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amr M. Saadawy
- Department of Diagnostic and Interventional Radiology and Molecular Imaging, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Heba Elsedfy
- Department of Medical Genetics, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
114
|
Yang S, Yang J, Zhao H, Deng R, Fan H, Zhang J, Yang Z, Zeng H, Kuang B, Shao L. The Protective Effects of γ-Tocotrienol on Muscle Stem Cells Through Inhibiting Reactive Oxidative Stress Production. Front Cell Dev Biol 2022; 10:820520. [PMID: 35372342 PMCID: PMC8965065 DOI: 10.3389/fcell.2022.820520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/01/2022] [Indexed: 11/25/2022] Open
Abstract
Pseudotrophic muscular dystrophy is a common clinical skeletal muscle necrotic disease, among which Duchenne muscular dystrophy (DMD) is the predominant. For such diseases, there is no clinically effective treatment, which is only symptomatic or palliative treatment. Oxidative stress and chronic inflammation are common pathological features of DMD. In recent years, it has been found that the pathophysiological changes of skeletal muscle in DMD mice are related to muscle stem cell failure. In the present study, we established a DMD mice model and provided tocotrienol (γ-tocotrienol, GT3), an antioxidant compound, to explore the relationship between the physiological state of muscle stem cells and oxidative stress. The results showed that the application of GT3 can reduce ROS production and cellular proliferation in the muscle stem cells of DMD mice, which is beneficial to promote the recovery of muscle stem cell function in DMD mice. GT3 treatment improved the differentiation ability of muscle stem cells in DMD mice with increasing numbers of MyoD+ cells. GT3 application significantly decreased percentages of CD45+ cells and PDGFRα+ fibro-adipogenic progenitors in the tibialis anterior of DMD mice, indicating that the increased inflammation and fibro-adipogenic progenitors were attenuated in GT3-treated DMD mice. These data suggest that increased ROS production causes dysfunctional muscle stem cell in DMD mice, which might provide a new avenue to treat DMD patients in the clinic.
Collapse
Affiliation(s)
- Shuo Yang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Juan Yang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Huiwen Zhao
- Department of Biological Genetics, School of Basic Medicine, Nanchang University, Nanchang, China
| | - Rong Deng
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Hancheng Fan
- Department of Histology and Embryology, School of Basic Medicine, Nanchang University, Nanchang, China
| | - Jinfu Zhang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Zihao Yang
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
| | - Huihong Zeng
- Department of Histology and Embryology, School of Basic Medicine, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, China
| | - Bohai Kuang
- Department of Biological Genetics, School of Basic Medicine, Nanchang University, Nanchang, China
| | - Lijian Shao
- Department of Occupational Health and Toxicology, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Interdisciplinary Science, Nanchang University, Nanchang, China
- *Correspondence: Lijian Shao,
| |
Collapse
|
115
|
Lingineni K, Aggarwal V, Morales JF, Conrado DJ, Corey D, Vong C, Burton J, Larkindale J, Romero K, Schmidt S, Kim S. Development of a model-based clinical trial simulation platform to optimize the design of clinical trials for Duchenne muscular dystrophy. CPT Pharmacometrics Syst Pharmacol 2022; 11:318-332. [PMID: 34877803 PMCID: PMC8923721 DOI: 10.1002/psp4.12753] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022] Open
Abstract
Early clinical trials of therapies to treat Duchenne muscular dystrophy (DMD), a fatal genetic X‐linked pediatric disease, have been designed based on the limited understanding of natural disease progression and variability in clinical measures over different stages of the continuum of the disease. The objective was to inform the design of DMD clinical trials by developing a disease progression model‐based clinical trial simulation (CTS) platform based on measures commonly used in DMD trials. Data were integrated from past studies through the Duchenne Regulatory Science Consortium founded by the Critical Path Institute (15 clinical trials and studies, 1505 subjects, 27,252 observations). Using a nonlinear mixed‐effects modeling approach, longitudinal dynamics of five measures were modeled (NorthStar Ambulatory Assessment, forced vital capacity, and the velocities of the following three timed functional tests: time to stand from supine, time to climb 4 stairs, and 10 meter walk‐run time). The models were validated on external data sets and captured longitudinal changes in the five measures well, including both early disease when function improves as a result of growth and development and the decline in function in later stages. The models can be used in the CTS platform to perform trial simulations to optimize the selection of inclusion/exclusion criteria, selection of measures, and other trial parameters. The data sets and models have been reviewed by the US Food and Drug Administration and the European Medicines Agency; have been accepted into the Fit‐for‐Purpose and Qualification for Novel Methodologies pathways, respectively; and will be submitted for potential endorsement by both agencies.
Collapse
Affiliation(s)
- Karthik Lingineni
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Diane Corey
- Critical Path Institute, Tucson, Arizona, USA
| | - Camille Vong
- Global Product Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | | | | | | | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | |
Collapse
|
116
|
Genetic Analysis of Forty MLPA-Negative Duchenne Muscular Dystrophy Patients by Whole-Exome Sequencing. J Mol Neurosci 2022; 72:1098-1107. [PMID: 35218518 DOI: 10.1007/s12031-022-01980-5] [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: 12/18/2021] [Accepted: 01/26/2022] [Indexed: 10/19/2022]
Abstract
This manuscript aimed to determine the underlying point mutations causing Duchenne muscular dystrophy (DMD) in a heterogeneous group of Iranian patients, who are clinically suspected. Whole-exome sequencing was utilized to detect disease-causing variants in 40 MLPA-negative DMD patients. Disease-causing variants were detected in the DMD gene in 36/40 of the patients (90%), and 4/40 of them (10%) remained undiagnosed. WES analysis revealed that nonsense variant was the most common type in our study (23/36 of the cases). Besides, 12/36 of the cases had frameshift variant, and one of the patients had a likely pathogenic splice variant in the DMD gene. Carrier testing revealed that 21/40 of the mothers had the identified variant. Therefore, most variants were inherited (58.3%), while 19/40 were de novo (41. 7%). The present study has demonstrated the importance of performing WES to detect disease-causing point mutations in MLPA-negative DMD patients and to identify carrier females. Due to regulatory challenges, the clinical development of therapeutic approaches is time-consuming and may not be available to all patients shortly. Therefore, it appears that the techniques used to accurately detect disease-causing variants in carrier mothers are a more efficient solution to prevent the increased prevalence of DMD.
Collapse
|
117
|
Morroni J, Schirone L, Valenti V, Zwergel C, Riera CS, Valente S, Vecchio D, Schiavon S, Ragno R, Mai A, Sciarretta S, Lozanoska-Ochser B, Bouchè M. Inhibition of PKCθ Improves Dystrophic Heart Phenotype and Function in a Novel Model of DMD Cardiomyopathy. Int J Mol Sci 2022; 23:ijms23042256. [PMID: 35216371 PMCID: PMC8880527 DOI: 10.3390/ijms23042256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Chronic cardiac muscle inflammation and subsequent fibrotic tissue deposition are key features in Duchenne Muscular Dystrophy (DMD). The treatment of choice for delaying DMD progression both in skeletal and cardiac muscle are corticosteroids, supporting the notion that chronic inflammation in the heart plays a pivotal role in fibrosis deposition and subsequent cardiac dysfunction. Nevertheless, considering the adverse effects associated with long-term corticosteroid treatments, there is a need for novel anti-inflammatory therapies. In this study, we used our recently described exercised mdx (ex mdx) mouse model characterised by accelerated heart pathology, and the specific PKCθ inhibitor Compound 20 (C20), to show that inhibition of this kinase leads to a significant reduction in the number of immune cells infiltrating the heart, as well as necrosis and fibrosis. Functionally, C20 treatment also prevented the reduction in left ventricle fractional shortening, which was typically observed in the vehicle-treated ex mdx mice. Based on these findings, we propose that PKCθ pharmacological inhibition could be an attractive therapeutic approach to treating dystrophic cardiomyopathy
Collapse
Affiliation(s)
- Jacopo Morroni
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Valentina Valenti
- Department of Cardiology, Ospedale Santa Maria Goretti, 04100 Latina, Italy;
| | - Clemens Zwergel
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Carles Sánchez Riera
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Daniele Vecchio
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Sonia Schiavon
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
| | - Rino Ragno
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185 Rome, Italy; (C.Z.); (S.V.); (R.R.); (A.M.)
| | - Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 00185 Rome, Italy; (L.S.); (D.V.); (S.S.); (S.S.)
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Biliana Lozanoska-Ochser
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
| | - Marina Bouchè
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy; (J.M.); (C.S.R.); (B.L.-O.)
- Correspondence:
| |
Collapse
|
118
|
Sergeeva OV, Shcherbinina EY, Shomron N, Zatsepin TS. Modulation of RNA Splicing by Oligonucleotides: Mechanisms of Action and Therapeutic Implications. Nucleic Acid Ther 2022; 32:123-138. [PMID: 35166605 DOI: 10.1089/nat.2021.0067] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of RNA splicing causes many diseases and disorders. Several therapeutic approaches have been developed to correct aberrant alternative splicing events for the treatment of cancers and hereditary diseases, including gene therapy and redirecting splicing, using small molecules or splice switching oligonucleotides (SSO). Significant advances in the chemistry and pharmacology of nucleic acid have led to the development of clinically approved SSO drugs for the treatment of spinal muscular dystrophy and Duchenne muscular dystrophy (DMD). In this review, we discuss the mechanisms of SSO action with emphasis on "less common" approaches to modulate alternative splicing, including bipartite and bifunctional SSO, oligonucleotide decoys for splice factors and SSO-mediated mRNA degradation via AS-NMD and NGD pathways. We briefly discuss the current progress and future perspectives of SSO therapy for rare and ultrarare diseases.
Collapse
Affiliation(s)
- Olga V Sergeeva
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | | | - Noam Shomron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Timofei S Zatsepin
- Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia.,Department of Chemistry, Moscow State University, Moscow, Russia
| |
Collapse
|
119
|
Bourg N, Vu Hong A, Lostal W, Jaber A, Guerchet N, Tanniou G, Bordier F, Bertil-Froidevaux E, Georger C, Daniele N, Richard I, Israeli D. Co-Administration of Simvastatin Does Not Potentiate the Benefit of Gene Therapy in the mdx Mouse Model for Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23042016. [PMID: 35216132 PMCID: PMC8878028 DOI: 10.3390/ijms23042016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common and cureless muscle pediatric genetic disease, which is caused by the lack or the drastically reduced expression of dystrophin. Experimental therapeutic approaches for DMD have been mainly focused in recent years on attempts to restore the expression of dystrophin. While significant progress was achieved, the therapeutic benefit of treated patients is still unsatisfactory. Efficiency in gene therapy for DMD is hampered not only by incompletely resolved technical issues, but likely also due to the progressive nature of DMD. It is indeed suspected that some of the secondary pathologies, which are evolving over time in DMD patients, are not fully corrected by the restoration of dystrophin expression. We recently identified perturbations of the mevalonate pathway and of cholesterol metabolism in DMD patients. Taking advantage of the mdx model for DMD, we then demonstrated that some of these perturbations are improved by treatment with the cholesterol-lowering drug, simvastatin. In the present investigation, we tested whether the combination of the restoration of dystrophin expression with simvastatin treatment could have an additive beneficial effect in the mdx model. We confirmed the positive effects of microdystrophin, and of simvastatin, when administrated separately, but detected no additive effect by their combination. Thus, the present study does not support an additive beneficial effect by combining dystrophin restoration with a metabolic normalization by simvastatin.
Collapse
Affiliation(s)
- Nathalie Bourg
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Ai Vu Hong
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - William Lostal
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Abbass Jaber
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Nicolas Guerchet
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Guillaume Tanniou
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Fanny Bordier
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Emilie Bertil-Froidevaux
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Christophe Georger
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Nathalie Daniele
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - Isabelle Richard
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
| | - David Israeli
- Généthon, 91000 Evry, France; (N.B.); (A.V.H.); (W.L.); (A.J.); (N.G.); (G.T.); (F.B.); (E.B.-F.); (C.G.); (N.D.); (I.R.)
- Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare Research Unit UMR_S951, 91000 Evry, France
- Correspondence: ; Tel.: + 33-1-6947-2967
| |
Collapse
|
120
|
Mahecha D, Nuñez H, Lattig MC, Duitama J. Machine Learning Models for Accurate Prioritization of Variants of Uncertain Significance. Hum Mutat 2022; 43:449-460. [PMID: 35143088 DOI: 10.1002/humu.24339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/04/2022] [Accepted: 01/23/2022] [Indexed: 11/08/2022]
Abstract
The growing use of next generation sequencing technologies on genetic diagnosis has produced an exponential increase in the number of Variants of Uncertain Significance (VUS). In this manuscript we compare three machine learning methods to classify VUS as Pathogenic or No pathogenic, implementing a Random Forest (RF), a Support Vector Machine (SVM), and a Multilayer Perceptron (MLP). To train the models, we extracted high quality variants from ClinVar that were previously classified as VUS. For each variant, we retrieved 9 conservation scores, the loss of function tool and allele frequencies. For the RF and SVM models, hyperparameters were tuned using cross validation with a grid search. The three models were tested on a non-overlapping set of variants that had been classified as VUS any time along the last three years but had been reclassified in august 2020. The three models yielded superior accuracy on this set compared to the benchmarked tools. The RF based model yielded the best performance across different variant types and was used to create VusPrize, an open source software tool for prioritization of variants of uncertain significance. We believe that our model can improve the process of genetic diagnosis in research and clinical settings. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Daniel Mahecha
- SIGEN, Alianza Universidad de los Andes - Fundación Santa Fe de Bogota, Colombia.,Systems and Computing Engineering Department, Universidad de los Andes, Colombia
| | - Haydemar Nuñez
- Systems and Computing Engineering Department, Universidad de los Andes, Colombia
| | - Maria C Lattig
- SIGEN, Alianza Universidad de los Andes - Fundación Santa Fe de Bogota, Colombia.,Facultad de Ciencias, Universidad de los Andes
| | - Jorge Duitama
- Systems and Computing Engineering Department, Universidad de los Andes, Colombia
| |
Collapse
|
121
|
Palazzo L, Pompele S, Rossi M, Rossi G, Spinoglio S, Testoni I. Facing Muscular Dystrophy During Covid-19 Pandemic: The Role of Support Associations and Spirituality. PASTORAL PSYCHOLOGY 2022; 71:217-231. [PMID: 35132281 PMCID: PMC8811345 DOI: 10.1007/s11089-022-00997-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 06/14/2023]
Abstract
Several researches in scientific literature analyze the theme of Muscular Dystrophy (MD), As well as many others focus on the theme of the Covid-19 pandemic; however, there is a rather limited number of studies that analyse how the pandemic has affected the life of people suffering from MD, especially during the time of the first lockdown in the spring of 2020. The present study has applied a qualitative research design with the aim to investigate how patients with MD have lived the social restrictions imposed for the contagion containment and whether the assistance of associations for their support has contributed to make the participants feel closer or more distant from the spiritual dimension. The analysis involved 12 participants, and they were presented with a semi-structured interview. The data obtained from the interviews have been analysed through a thematic analysis from which 4 thematic areas have emerged: (1) the impact of the pandemic on an emotional level; (2) the illness management and the role of family; (3) the role of the associations; (4) aspects related to spirituality. The crucial role that the closeness of family and the activities promoted remotely by the associations for patients' support has emerged, since they have allowed the participants to feel united by something beyond, to discover new aspects of themselves, to give more value to Life and to move closer to their spiritual dimension.
Collapse
Affiliation(s)
- Lorenza Palazzo
- Department of Philosophy, Sociology, Pedagogy and Applied Psychology (FISPPA), University of Padova, 35131 Padova, Italy
| | - Sara Pompele
- Department of Philosophy, Sociology, Pedagogy and Applied Psychology (FISPPA), University of Padova, 35131 Padova, Italy
| | - Marta Rossi
- Department of Philosophy, Sociology, Pedagogy and Applied Psychology (FISPPA), University of Padova, 35131 Padova, Italy
| | - Gabriella Rossi
- Unione Italiana Lotta Alla Distrofia Muscolare (UILDM) - “G. Bergna” section of Monza and Brianza, 20900 Monza e Brianza (MB), Italy
| | - Simona Spinoglio
- Unione Italiana Lotta Alla Distrofia Muscolare (UILDM) - “G. Bergna” section of Monza and Brianza, 20900 Monza e Brianza (MB), Italy
| | - Ines Testoni
- Department of Philosophy, Sociology, Pedagogy and Applied Psychology (FISPPA), University of Padova, 35131 Padova, Italy
- Emili Sagol Creative Arts Therapies Research Center, University of Haifa, 3498838 Haifa, Israel
| |
Collapse
|
122
|
Matsuzaka Y, Hirai Y, Hashido K, Okada T. Therapeutic Application of Extracellular Vesicles-Capsulated Adeno-Associated Virus Vector via nSMase2/Smpd3, Satellite, and Immune Cells in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:1551. [PMID: 35163475 PMCID: PMC8836108 DOI: 10.3390/ijms23031551] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by loss-of-function mutations in the dystrophin gene on chromosome Xp21. Disruption of the dystrophin-glycoprotein complex (DGC) on the cell membrane causes cytosolic Ca2+ influx, resulting in protease activation, mitochondrial dysfunction, and progressive myofiber degeneration, leading to muscle wasting and fragility. In addition to the function of dystrophin in the structural integrity of myofibers, a novel function of asymmetric cell division in muscular stem cells (satellite cells) has been reported. Therefore, it has been suggested that myofiber instability may not be the only cause of dystrophic degeneration, but rather that the phenotype might be caused by multiple factors, including stem cell and myofiber functions. Furthermore, it has been focused functional regulation of satellite cells by intracellular communication of extracellular vesicles (EVs) in DMD pathology. Recently, a novel molecular mechanism of DMD pathogenesis-circulating RNA molecules-has been revealed through the study of target pathways modulated by the Neutral sphingomyelinase2/Neutral sphingomyelinase3 (nSMase2/Smpd3) protein. In addition, adeno-associated virus (AAV) has been clinically applied for DMD therapy owing to the safety and long-term expression of transduction genes. Furthermore, the EV-capsulated AAV vector (EV-AAV) has been shown to be a useful tool for the intervention of DMD, because of the high efficacy of the transgene and avoidance of neutralizing antibodies. Thus, we review application of AAV and EV-AAV vectors for DMD as novel therapeutic strategy.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan;
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8551, Japan;
| | - Yukihiko Hirai
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan;
| | - Kazuo Hashido
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo 187-8551, Japan;
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, University of Tokyo, Minato-ku, Tokyo 108-8639, Japan;
| |
Collapse
|
123
|
Lee J, An S, Lee SJ, Kang JS. Protein Arginine Methyltransferases in Neuromuscular Function and Diseases. Cells 2022; 11:364. [PMID: 35159176 PMCID: PMC8834056 DOI: 10.3390/cells11030364] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Neuromuscular diseases (NMDs) are characterized by progressive loss of muscle mass and strength that leads to impaired body movement. It not only severely diminishes the quality of life of the patients, but also subjects them to increased risk of secondary medical conditions such as fall-induced injuries and various chronic diseases. However, no effective treatment is currently available to prevent or reverse the disease progression. Protein arginine methyltransferases (PRMTs) are emerging as a potential therapeutic target for diverse diseases, such as cancer and cardiovascular diseases. Their expression levels are altered in the patients and molecular mechanisms underlying the association between PRMTs and the diseases are being investigated. PRMTs have been shown to regulate development, homeostasis, and regeneration of both muscle and neurons, and their association to NMDs are emerging as well. Through inhibition of PRMT activities, a few studies have reported suppression of cytotoxic phenotypes observed in NMDs. Here, we review our current understanding of PRMTs' involvement in the pathophysiology of NMDs and potential therapeutic strategies targeting PRMTs to address the unmet medical need.
Collapse
Affiliation(s)
- Jinwoo Lee
- Research Institute for Aging-Related Diseases, AniMusCure Inc., Suwon 16419, Korea;
| | - Subin An
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Korea;
- Single Cell Network Research Center, Sungkyunkwan University, Suwon 16419, Korea
| | - Sang-Jin Lee
- Research Institute for Aging-Related Diseases, AniMusCure Inc., Suwon 16419, Korea;
| | - Jong-Sun Kang
- Department of Molecular Cell Biology, School of Medicine, Sungkyunkwan University, Suwon 16419, Korea;
- Single Cell Network Research Center, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
124
|
Duelen R, Costamagna D, Gilbert G, Waele LD, Goemans N, Desloovere K, Verfaillie CM, Sipido KR, Buyse GM, Sampaolesi M. Human iPSC model reveals a central role for NOX4 and oxidative stress in Duchenne cardiomyopathy. Stem Cell Reports 2022; 17:352-368. [PMID: 35090586 PMCID: PMC8828550 DOI: 10.1016/j.stemcr.2021.12.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disorder caused by mutations in the Dystrophin gene. Cardiomyopathy is a major cause of early death. We used DMD-patient-specific human induced pluripotent stem cells (hiPSCs) to model cardiomyopathic features and unravel novel pathologic insights. Cardiomyocytes (CMs) differentiated from DMD hiPSCs showed enhanced premature cell death due to significantly elevated intracellular reactive oxygen species (ROS) resulting from depolarized mitochondria and increased NADPH oxidase 4 (NOX4). CRISPR-Cas9 correction of Dystrophin restored normal ROS levels. ROS reduction by N-acetyl-L-cysteine (NAC), ataluren (PTC124), and idebenone improved hiPSC-CM survival. We show that oxidative stress in DMD hiPSC-CMs was counteracted by stimulating adenosine triphosphate (ATP) production. ATP can bind to NOX4 and partially inhibit the ROS production. Considering the complexity and the early cellular stress responses in DMD cardiomyopathy, we propose targeting ROS production and preventing detrimental effects of NOX4 on DMD CMs as promising therapeutic strategy.
Collapse
Affiliation(s)
- Robin Duelen
- Translational Cardiomyology Lab, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Herestraat 49 - O&N4 - bus 804, 3000 Leuven, Belgium
| | - Domiziana Costamagna
- Translational Cardiomyology Lab, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Herestraat 49 - O&N4 - bus 804, 3000 Leuven, Belgium; Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Guillaume Gilbert
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Liesbeth De Waele
- Pediatric Neurology, University Hospitals Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Nathalie Goemans
- Pediatric Neurology, University Hospitals Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Kaat Desloovere
- Research Group for Neurorehabilitation, Department of Rehabilitation Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Catherine M Verfaillie
- Stem Cell Institute Leuven, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Karin R Sipido
- Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Gunnar M Buyse
- Pediatric Neurology, University Hospitals Leuven, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | - Maurilio Sampaolesi
- Translational Cardiomyology Lab, Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Herestraat 49 - O&N4 - bus 804, 3000 Leuven, Belgium; Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy.
| |
Collapse
|
125
|
Angelini G, Mura G, Messina G. Therapeutic approaches to preserve the musculature in Duchenne Muscular Dystrophy: The importance of the secondary therapies. Exp Cell Res 2022; 410:112968. [PMID: 34883113 DOI: 10.1016/j.yexcr.2021.112968] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/15/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies (MDs) are heterogeneous diseases, characterized by primary wasting of skeletal muscle, which in severe cases, such as Duchenne Muscular Dystrophy (DMD), leads to wheelchair dependency, respiratory failure, and premature death. Research is ongoing to develop efficacious therapies, particularly for DMD. Most of the efforts, currently focusing on correcting or restoring the primary defect of MDs, are based on gene-addition, exon-skipping, stop codon read-through, and genome-editing. Although promising, most of them revealed several practical limitations. Shared knowledge in the field is that, in order to be really successful, any therapeutic approach has to rely on spared functional muscle tissue, restricting the number of patients eligible for clinical trials to the youngest and less compromised individuals. In line with this, many therapeutic strategies aim to preserve muscle tissue and function. This Review outlines the most interesting and recent studies addressing the secondary outcomes of DMD and how to better deliver the therapeutic agents. In the future, the effective treatment of DMD will likely require combinations of therapies addressing both the primary genetic defect and its consequences.
Collapse
Affiliation(s)
- Giuseppe Angelini
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
126
|
Jablonka S, Hennlein L, Sendtner M. Therapy development for spinal muscular atrophy: perspectives for muscular dystrophies and neurodegenerative disorders. Neurol Res Pract 2022; 4:2. [PMID: 34983696 PMCID: PMC8725368 DOI: 10.1186/s42466-021-00162-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/21/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Major efforts have been made in the last decade to develop and improve therapies for proximal spinal muscular atrophy (SMA). The introduction of Nusinersen/Spinraza™ as an antisense oligonucleotide therapy, Onasemnogene abeparvovec/Zolgensma™ as an AAV9-based gene therapy and Risdiplam/Evrysdi™ as a small molecule modifier of pre-mRNA splicing have set new standards for interference with neurodegeneration. MAIN BODY Therapies for SMA are designed to interfere with the cellular basis of the disease by modifying pre-mRNA splicing and enhancing expression of the Survival Motor Neuron (SMN) protein, which is only expressed at low levels in this disorder. The corresponding strategies also can be applied to other disease mechanisms caused by loss of function or toxic gain of function mutations. The development of therapies for SMA was based on the use of cell culture systems and mouse models, as well as innovative clinical trials that included readouts that had originally been introduced and optimized in preclinical studies. This is summarized in the first part of this review. The second part discusses current developments and perspectives for amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease, as well as the obstacles that need to be overcome to introduce RNA-based therapies and gene therapies for these disorders. CONCLUSION RNA-based therapies offer chances for therapy development of complex neurodegenerative disorders such as amyotrophic lateral sclerosis, muscular dystrophies, Parkinson's and Alzheimer's disease. The experiences made with these new drugs for SMA, and also the experiences in AAV gene therapies could help to broaden the spectrum of current approaches to interfere with pathophysiological mechanisms in neurodegeneration.
Collapse
Affiliation(s)
- Sibylle Jablonka
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| | - Luisa Hennlein
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital of Wuerzburg, Versbacher Str. 5, 97078, Wuerzburg, Germany.
| |
Collapse
|
127
|
Morris CE, Wheeler JJ, Joos B. The Donnan-dominated resting state of skeletal muscle fibers contributes to resilience and longevity in dystrophic fibers. J Gen Physiol 2022; 154:212743. [PMID: 34731883 PMCID: PMC8570295 DOI: 10.1085/jgp.202112914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/30/2021] [Indexed: 11/28/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked dystrophin-minus muscle-wasting disease. Ion homeostasis in skeletal muscle fibers underperforms as DMD progresses. But though DMD renders these excitable cells intolerant of exertion, sodium overloaded, depolarized, and spontaneously contractile, they can survive for several decades. We show computationally that underpinning this longevity is a strikingly frugal, robust Pump-Leak/Donnan (P-L/D) ion homeostatic process. Unlike neurons, which operate with a costly “Pump-Leak–dominated” ion homeostatic steady state, skeletal muscle fibers operate with a low-cost “Donnan-dominated” ion homeostatic steady state that combines a large chloride permeability with an exceptionally small sodium permeability. Simultaneously, this combination keeps fiber excitability low and minimizes pump expenditures. As mechanically active, long-lived multinucleate cells, skeletal muscle fibers have evolved to handle overexertion, sarcolemmal tears, ischemic bouts, etc.; the frugality of their Donnan dominated steady state lets them maintain the outsized pump reserves that make them resilient during these inevitable transient emergencies. Here, P-L/D model variants challenged with DMD-type insult/injury (low pump-strength, overstimulation, leaky Nav and cation channels) show how chronic “nonosmotic” sodium overload (observed in DMD patients) develops. Profoundly severe DMD ion homeostatic insult/injury causes spontaneous firing (and, consequently, unwanted excitation–contraction coupling) that elicits cytotoxic swelling. Therefore, boosting operational pump-strength and/or diminishing sodium and cation channel leaks should help extend DMD fiber longevity.
Collapse
Affiliation(s)
- Catherine E Morris
- Neuroscience, Ottawa Hospital Research Institute, Ottawa, Canada.,Center for Neural Dynamics, University of Ottawa, Ottawa, Canada
| | | | - Béla Joos
- Center for Neural Dynamics, University of Ottawa, Ottawa, Canada.,Department of Physics, University of Ottawa, Ottawa, Canada
| |
Collapse
|
128
|
Kong L, Li S, Zhao Z, Feng J, Chen G, Liu L, Tang W, Li S, Li F, Han X, Wu D, Zhang H, Sun L, Kong X. Haplotype-Based Noninvasive Prenatal Diagnosis of 21 Families With Duchenne Muscular Dystrophy: Real-World Clinical Data in China. Front Genet 2022; 12:791856. [PMID: 34970304 PMCID: PMC8712857 DOI: 10.3389/fgene.2021.791856] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
Noninvasive prenatal diagnosis (NIPD) of single-gene disorders has recently become the focus of clinical laboratories. However, reports on the clinical application of NIPD of Duchenne muscular dystrophy (DMD) are limited. This study aimed to evaluate the detection performance of haplotype-based NIPD of DMD in a real clinical environment. Twenty-one DMD families at 7-12 weeks of gestation were prospectively recruited. DNA libraries of cell-free DNA from the pregnant and genomic DNA from family members were captured using a custom assay for the enrichment of DMD gene exons and spanning single-nucleotide polymorphisms, followed by next-generation sequencing. Parental haplotype phasing was based on family linkage analysis, and fetal genotyping was inferred using the Bayes factor through target maternal plasma sequencing. Finally, the entire experimental process was promoted in the local clinical laboratory. We recruited 13 complete families, 6 families without paternal samples, and 2 families without probands in which daughter samples were collected. Two different maternal haplotypes were constructed based on family members in all 21 pedigrees at as early as 7 gestational weeks. Among the included families, the fetal genotypes of 20 families were identified at the first blood collection, and a second blood collection was performed for another family due to low fetal concentration. The NIPD result of each family was reported within 1 week. The fetal fraction in maternal cfDNA ranged from 1.87 to 11.68%. In addition, recombination events were assessed in two fetuses. All NIPD results were concordant with the findings of invasive prenatal diagnosis (chorionic villus sampling or amniocentesis). Exon capture and haplotype-based NIPD of DMD are regularly used for DMD genetic diagnosis, carrier screening, and noninvasive prenatal diagnosis in the clinic. Our method, haplotype-based early screening for DMD fetal genotyping via cfDNA sequencing, has high feasibility and accuracy, a short turnaround time, and is inexpensive in a real clinical environment.
Collapse
Affiliation(s)
- Lingrong Kong
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.,Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaojun Li
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Zhenhua Zhao
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Feng
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Guangquan Chen
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lina Liu
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiqin Tang
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Suqing Li
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Feifei Li
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Xiujuan Han
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Di Wu
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Haichuan Zhang
- Celula (China) Medical Technology Co., Ltd., Chengdu, China
| | - Luming Sun
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiangdong Kong
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
129
|
Ritso M, Tung LW, Rossi FMV. Emerging skeletal muscle stromal cell diversity: Functional divergence in fibro/adipogenic progenitor and mural cell populations. Exp Cell Res 2022; 410:112947. [PMID: 34822813 DOI: 10.1016/j.yexcr.2021.112947] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/15/2021] [Accepted: 11/21/2021] [Indexed: 12/27/2022]
Abstract
While the majority of healthy skeletal muscle consists of multinucleated syncytial repetitive contractile myofibers, repaired by skeletal muscle stem cells when damaged, the maintenance of muscle function also requires a range of tissue-resident stromal populations. In fact, the careful orchestration of damage response processes upon muscle injury relies heavily on stromal cell contribution for effective repair. The two main types of muscle-resident stromal cells are fibro/adipogenic progenitors and mural cells. The latter is comprised of pericytes and vascular smooth muscle cells. Recent publications identifying common markers for stromal cell populations have allowed investigating population dynamics throughout the regenerative process at a higher resolution. Mounting evidence now suggests that subpopulations with distinct roles may exist among stromal cells. In various degenerative muscle wasting conditions, critical cross-talk and spatial signalling amongst various cell populations become dysregulated. This can result in the failure to curb pathological fibro/adipogenic progenitor proliferation and propensity for laying down excessive extracellular matrix, which in turn leads to fibrotic infiltration, reduced contractile units and gradual decline in muscle function. Restoration of physiologically appropriate stromal cell function is therefore just as crucial for therapeutic targeting as the homeostatic maintenance of muscle function.
Collapse
Affiliation(s)
- Morten Ritso
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Fabio M V Rossi
- School of Biomedical Engineering, 2222 Health Sciences Mall, The University of British Columbia, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
130
|
Ricci G, Bello L, Torri F, Schirinzi E, Pegoraro E, Siciliano G. Therapeutic opportunities and clinical outcome measures in Duchenne muscular dystrophy. Neurol Sci 2022; 43:625-633. [PMID: 35608735 PMCID: PMC9126754 DOI: 10.1007/s10072-022-06085-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/14/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a devastatingly severe genetic muscle disease characterized by childhood-onset muscle weakness, leading to loss of motor function and premature death due to respiratory and cardiac insufficiency. DISCUSSION In the following three and half decades, DMD kept its paradigmatic role in the field of muscle diseases, with first systematic description of disease progression with ad hoc outcome measures and the first attempts at correcting the disease-causing gene defect by several molecular targets. Clinical trials are critical for developing and evaluating new treatments for DMD. CONCLUSIONS In the last 20 years, research efforts converged in characterization of the disease mechanism and development of therapeutic strategies. Same effort needs to be dedicated to the development of outcome measures able to capture clinical benefit in clinical trials.
Collapse
Affiliation(s)
- Giulia Ricci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Francesca Torri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Erika Schirinzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
131
|
Roshmi RR, Yokota T. Pharmacological Profile of Viltolarsen for the Treatment of Duchenne Muscular Dystrophy: A Japanese Experience. Clin Pharmacol 2021; 13:235-242. [PMID: 34938127 PMCID: PMC8688746 DOI: 10.2147/cpaa.s288842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/12/2021] [Indexed: 01/11/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal, X-linked recessive disorder characterized by progressive muscle loss and cardiorespiratory complications. Mutations in the DMD gene that eliminate the production of dystrophin protein are the underlying causes of DMD. Viltolarsen is a drug of phosphorodiamidate morpholino oligomer (PMO) chemistry, designed to skip exon 53 of the DMD gene. It aims to produce truncated but partially functional dystrophin in DMD patients and restore muscle function. Based on a preclinical study showing the ability of antisense PMOs targeting the DMD gene to improve muscle function in a large animal model, viltolarsen was developed by Nippon Shinyaku and the National Center of Neurology and Psychiatry in Japan. Following clinical trials conducted in Japan, Canada, and the United States showing significant improvements in muscle function, viltolarsen was approved for medical use in Japan in March 2020 and the United States in August 2020, respectively. Viltolarsen is a mutation-specific drug and will work for 8% of the persons with DMD who carry mutations amenable to exon 53 skipping. This review summarizes the pharmacological profile of viltolarsen, important clinical trials, and challenges, focusing on the contribution of Japanese patients and researchers in its development.
Collapse
Affiliation(s)
- Rohini Roy Roshmi
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
132
|
Creisméas A, Gazaille C, Bourdon A, Lallemand MA, François V, Allais M, Ledevin M, Larcher T, Toumaniantz G, Lafoux A, Huchet C, Anegon I, Adjali O, Le Guiner C, Fraysse B. TRPC3, but not TRPC1, as a good therapeutic target for standalone or complementary treatment of DMD. J Transl Med 2021; 19:519. [PMID: 34930315 PMCID: PMC8686557 DOI: 10.1186/s12967-021-03191-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked inherited disease caused by mutations in the gene encoding dystrophin that leads to a severe and ultimately life limiting muscle-wasting condition. Recombinant adeno-associated vector (rAAV)-based gene therapy is promising, but the size of the full-length dystrophin cDNA exceeds the packaging capacity of a rAAV. Alternative or complementary strategies that could treat DMD patients are thus needed. Intracellular calcium overload due to a sarcolemma permeability to calcium (SPCa) increase is an early and critical step of the DMD pathogenesis. We assessed herein whether TRPC1 and TRPC3 calcium channels may be involved in skeletal muscle SPCa alterations and could represent therapeutic targets to treat DMD. Methods All experiments were conducted in the DMDmdx rat, an animal model that closely reproduces the human DMD disease. We measured the cytosolic calcium concentration ([Ca2+]c) and SPCa in EDL (Extensor Digitorum Longus) muscle fibers from age-matched WT and DMDmdx rats of 1.5 to 7 months old. TRPC1 and TRPC3 expressions were measured in the EDL muscles at both the mRNA and protein levels, by RT-qPCR, western blot and immunocytofluorescence analysis. Results As expected from the malignant hyperthermia like episodes observed in several DMDmdx rats, calcium homeostasis alterations were confirmed by measurements of early increases in [Ca2+]c and SPCa in muscle fibers. TRPC3 and TRPC1 protein levels were increased in DMDmdx rats. This was observed as soon as 1.5 months of age for TRPC3 but only at 7 months of age for TRPC1. A slight but reliable shift of the TRPC3 apparent molecular weight was observed in DMDmdx rat muscles. Intracellular localization of both channels was not altered. We thus focused our attention on TRPC3. Application of Pyr10, a specific inhibitor of TRPC3, abolished the differences between SPCa values measured in WT and DMDmdx. Finally, we showed that a rAAV-microdystrophin based treatment induced a high microdystrophin expression but only partial prevention of calcium homeostasis alterations, skeletal muscle force and TRPC3 protein increase. Conclusions All together our results show that correcting TRPC3 channel expression and/or activity appear to be a promising approach as a single or as a rAAV-based complementary therapy to treat DMD.
Collapse
Affiliation(s)
- Anna Creisméas
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Claire Gazaille
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Audrey Bourdon
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Marc-Antoine Lallemand
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Virginie François
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Marine Allais
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | | | | | - Gilles Toumaniantz
- L'Institut du Thorax, Université de Nantes, CNRS, INSERM UMR 1087, Nantes, France
| | - Aude Lafoux
- Therassay Platform, Capacités, Université de Nantes, Nantes, France
| | - Corinne Huchet
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Ignacio Anegon
- INSERM, UMR 1064-Center for Research in Transplantation and Immunology, ITUN, CHU Nantes, Université de Nantes, Faculté de Médecine, Nantes, France
| | - Oumeya Adjali
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Caroline Le Guiner
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France
| | - Bodvaël Fraysse
- Nantes Gene Therapy Laboratory, Université de Nantes, INSERM UMR 1089, IRS 2 Nantes Biotech, CHU de Nantes, 22, Boulevard Bénoni Goullin, 44200, Nantes, France.
| |
Collapse
|
133
|
Petrof BJ. Macrophage plasticity in Duchenne muscular dystrophy: a nexus of pathological remodelling with therapeutic implications. J Physiol 2021; 600:3455-3464. [PMID: 34904234 DOI: 10.1113/jp281673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/03/2021] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by chronic skeletal muscle necrosis, leading to muscle regeneration failure and fibrosis. Although macrophages (MPs) are normally essential for muscle regeneration, dysregulated MP function promotes pathological muscle remodelling. Infiltrating MPs can be predominantly pro-inflammatory (M1 biased), anti-inflammatory (M2 biased) or of a mixed phenotype and can originate from the adult bone marrow (monocyte dependent) or embryonic precursors (monocyte independent). In mdx mice (genetic model of DMD) lacking either Toll-like receptor (Tlr) 2 or Tlr4, it is found that MP infiltration of dystrophic muscle is significantly reduced and that the MP phenotype is shifted toward a more anti-inflammatory profile. This is accompanied by significant improvements in muscle histology and force production. Lack of the chemokine receptor CCR2, which impedes monocyte release from the bone marrow, leads to similar beneficial effects in mdx mice. Evidence was also found for Tlr4-regulated induction of trained innate immunity in MPs cultured from the bone marrow of mdx mice before their entry into the muscle. These MPs exhibit epigenetic and metabolic alterations, accompanied by non-specific hyper-responsiveness to multiple stimuli, which is manifested by potentiated upregulation of both pro- and anti-inflammatory genes. In summary, exaggerated recruitment of monocyte-derived MPs and signs of trained innate immunity at the level of the bone marrow are features of the immunophenotype associated with dystrophic muscle disease. These phenomena are regulated by Toll-like receptors that bind endogenous damage-associated molecular pattern (DAMP) molecules, suggesting that DAMP release from dystrophic muscles modulates MP plasticity at the bone marrow level through Toll-like receptor-driven mechanisms.
Collapse
Affiliation(s)
- Basil J Petrof
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Respiratory Division, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
134
|
A Dystrophin Exon-52 Deleted Miniature Pig Model of Duchenne Muscular Dystrophy and Evaluation of Exon Skipping. Int J Mol Sci 2021; 22:ijms222313065. [PMID: 34884867 PMCID: PMC8657897 DOI: 10.3390/ijms222313065] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disorder caused by mutations in the DMD gene and the subsequent lack of dystrophin protein. Recently, phosphorodiamidate morpholino oligomer (PMO)-antisense oligonucleotides (ASOs) targeting exon 51 or 53 to reestablish the DMD reading frame have received regulatory approval as commercially available drugs. However, their applicability and efficacy remain limited to particular patients. Large animal models and exon skipping evaluation are essential to facilitate ASO development together with a deeper understanding of dystrophinopathies. Using recombinant adeno-associated virus-mediated gene targeting and somatic cell nuclear transfer, we generated a Yucatan miniature pig model of DMD with an exon 52 deletion mutation equivalent to one of the most common mutations seen in patients. Exon 52-deleted mRNA expression and dystrophin deficiency were confirmed in the skeletal and cardiac muscles of DMD pigs. Accordingly, dystrophin-associated proteins failed to be recruited to the sarcolemma. The DMD pigs manifested early disease onset with severe bodywide skeletal muscle degeneration and with poor growth accompanied by a physical abnormality, but with no obvious cardiac phenotype. We also demonstrated that in primary DMD pig skeletal muscle cells, the genetically engineered exon-52 deleted pig DMD gene enables the evaluation of exon 51 or 53 skipping with PMO and its advanced technology, peptide-conjugated PMO. The results show that the DMD pigs developed here can be an appropriate large animal model for evaluating in vivo exon skipping efficacy.
Collapse
|
135
|
Aoki Y, Rocha CSJ, Lehto T, Miyatake S, Johansson H, Hashimoto Y, Nordin JZ, Mager I, Aoki M, Graham M, Sathyaprakash C, Roberts TC, Wood MJA, Behlke MA, Andaloussi SE. Fine Tuning of Phosphorothioate Inclusion in 2'-O-Methyl Oligonucleotides Contributes to Specific Cell Targeting for Splice-Switching Modulation. Front Physiol 2021; 12:689179. [PMID: 34721051 PMCID: PMC8548633 DOI: 10.3389/fphys.2021.689179] [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: 03/31/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Splice-switching antisense oligonucleotide- (SSO-) mediated correction of framedisrupting mutation-containing premessenger RNA (mRNA) transcripts using exon skipping is a highly promising treatment method for muscular diseases such as Duchenne muscular dystrophy (DMD). Phosphorothioate (PS) chemistry, a commonly used oligonucleotide modification, has been shown to increase the stability of and improve the pharmacokinetics of SSOs. However, the effect of PS inclusion in 2′-O-methyl SSOs (2OMe) on cellular uptake and splice switching is less well-understood. At present, we demonstrate that the modification of PS facilitates the uptake of 2OMe in H2k-mdx myoblasts. Furthermore, we found a dependency of SSO nuclear accumulation and high splice-switching activity on PS inclusion in 2OMe (2OMePS), as tested in various reporter cell lines carrying pLuc/705. Increased exon-inclusion activity was observed in muscle, neuronal, liver, and bone cell lineages via both the gymnotic uptake and lipofection of 2OMePS. Using the photoactivatable ribonucleoside-enhanced crosslinking and a subsequent proteomic approach, we identified several 2OMePS-binding proteins, which are likely to play a role in the trafficking of 2OMePS to the nucleus. Ablation of one of them, Ncl by small-interfering RNA (siRNA) enhanced 2OMePS uptake in C2C12 myoblasts and upregulated luciferase RNA splicing in the HeLa Luc/705 reporter cell line. Overall, we demonstrate that PS inclusion increases nuclear delivery and splice switching in muscle, neuronal, liver, and bone cell lineages and that the modulation of 2OMePS-binding partners may improve SSO delivery.
Collapse
Affiliation(s)
- Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan.,Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Cristina S J Rocha
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Taavi Lehto
- Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Shouta Miyatake
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Henrik Johansson
- Department of Oncology-Pathology, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Yasumasa Hashimoto
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Joel Z Nordin
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan.,Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| | - Imre Mager
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Misako Aoki
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - McClorey Graham
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Chaitra Sathyaprakash
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Thomas C Roberts
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Matthew J A Wood
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Mark A Behlke
- Integrated DNA Technologies, Inc., Coralville, IA, United States
| | - Samir El Andaloussi
- Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,Department of Laboratory Medicine, Clinical Research Center, Karolinska Institutet, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
136
|
Servais L, Camino E, Clement A, McDonald CM, Lukawy J, Lowes LP, Eggenspieler D, Cerreta F, Strijbos P. First Regulatory Qualification of a Novel Digital Endpoint in Duchenne Muscular Dystrophy: A Multi-Stakeholder Perspective on the Impact for Patients and for Drug Development in Neuromuscular Diseases. Digit Biomark 2021; 5:183-190. [PMID: 34723071 DOI: 10.1159/000517411] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background Functional outcome measures used to assess efficacy in clinical trials of investigational treatments for rare neuromuscular diseases like Duchenne muscular dystrophy (DMD) are performance-based tasks completed by the patient during hospital visits. These are prone to bias and may not reflect motor abilities in real-world settings. Digital tools, such as wearable devices and other remote sensors, provide the opportunity for continuous, objective, and sensitive measurements of functional ability during daily life. Maintaining ambulation is of key importance to individuals with DMD. Stride velocity 95th centile (SV95C) is the first wearable acquired digital endpoint to receive qualification from the European Medicines Agency (EMA) to quantify the ambulation ability of ambulant DMD patients aged ≥5 years in drug therapeutic studies; it is also currently under review for the US Food and Drug Administration (FDA) qualification. Summary Focusing on SV95C as a key example, we describe perspectives of multiple stakeholders on the promise of novel digital endpoints in neuromuscular disease drug development.
Collapse
Affiliation(s)
- Laurent Servais
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, United Kingdom.,Division of Child Neurology, Centre de Référence des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège and University of Liège, Liege, Belgium
| | - Eric Camino
- Parent Project Muscular Dystrophy, Hackensack, New Jersey, USA
| | | | - Craig M McDonald
- University of California Davis Health, Sacramento, California, USA
| | | | - Linda P Lowes
- Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | | | | | | |
Collapse
|
137
|
Rodríguez-Fdez S, Bustelo XR. Rho GTPases in Skeletal Muscle Development and Homeostasis. Cells 2021; 10:cells10112984. [PMID: 34831205 PMCID: PMC8616218 DOI: 10.3390/cells10112984] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Rho guanosine triphosphate hydrolases (GTPases) are molecular switches that cycle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state during signal transduction. As such, they regulate a wide range of both cellular and physiological processes. In this review, we will summarize recent work on the role of Rho GTPase-regulated pathways in skeletal muscle development, regeneration, tissue mass homeostatic balance, and metabolism. In addition, we will present current evidence that links the dysregulation of these GTPases with diseases caused by skeletal muscle dysfunction. Overall, this information underscores the critical role of a number of members of the Rho GTPase subfamily in muscle development and the overall metabolic balance of mammalian species.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence: or
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
138
|
Hakim CH, Yang HT, Burke MJ, Teixeira J, Jenkins GJ, Yang NN, Yao G, Duan D. Extensor carpi ulnaris muscle shows unexpected slow-to-fast fiber type switch in Duchenne muscular dystrophy dogs. Dis Model Mech 2021; 14:273743. [PMID: 34704592 PMCID: PMC8688408 DOI: 10.1242/dmm.049006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022] Open
Abstract
Aged dystrophin-null canines are excellent models for studying experimental therapies for Duchenne muscular dystrophy, a lethal muscle disease caused by dystrophin deficiency. To establish the baseline, we studied the extensor carpi ulnaris (ECU) muscle in 15 terminal age (3-year-old) male affected dogs and 15 age/sex-matched normal dogs. Affected dogs showed histological and anatomical hallmarks of dystrophy, including muscle inflammation and fibrosis, myofiber size variation and centralized myonuclei, as well as a significant reduction of muscle weight, muscle-to-body weight ratio and muscle cross-sectional area. To rigorously characterize the contractile properties of the ECU muscle, we developed a novel in situ assay. Twitch and tetanic force, contraction and relaxation rate, and resistance to eccentric contraction-induced force loss were significantly decreased in affected dogs. Intriguingly, the time-to-peak tension and half-relaxation time were significantly shortened in affected dogs. Contractile kinetics predicted an unforeseen slow-to-fast myofiber-type switch, which we confirmed at the protein and transcript level. Our study establishes a foundation for studying long-term and late-stage therapeutic interventions in dystrophic canines. The unexpected myofiber-type switch highlights the complexity of muscle remodeling in dystrophic large mammals. This article has an associated First Person interview with the first author of the paper. Summary: A slow-to-fast fiber-type switch in dystrophic canine ECU muscle is revealed by contraction kinetics and myosin protein and transcript expression. This highlights the complexity of muscle remodeling in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA.,National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Hsiao T Yang
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA
| | - Matthew J Burke
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA
| | - James Teixeira
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA
| | - Gregory J Jenkins
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA
| | - N N Yang
- National Center for Advancing Translational Sciences, NIH, Bethesda, MD, USA
| | - Gang Yao
- Department of Biomedical, Biological & Chemical Engineering, College of Engineering, The University of Missouri, Columbia, MO, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, The University of Missouri, Columbia, MO, USA.,Department of Biomedical, Biological & Chemical Engineering, College of Engineering, The University of Missouri, Columbia, MO, USA.,Department of Neurology, School of Medicine, The University of Missouri, Columbia, MO, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, The University of Missouri, Columbia, MO, USA
| |
Collapse
|
139
|
Naarding KJ, van der Holst M, van Zwet EW, van de Velde NM, de Groot IJM, Verschuuren JJGM, Kan HE, Niks EH. Association of Elbow Flexor MRI Fat Fraction With Loss of Hand-to-Mouth Movement in Patients With Duchenne Muscular Dystrophy. Neurology 2021; 97:e1737-e1742. [PMID: 34493619 PMCID: PMC8605612 DOI: 10.1212/wnl.0000000000012724] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVES To study the potential of quantitative MRI (qMRI) fat fraction (FF) as a biomarker in nonambulant patients with Duchenne muscular dystrophy (DMD), we assessed the additive predictive value of elbow flexor FF to age at loss of hand-to-mouth movement. METHODS Nonambulant patients with DMD (age ≥8 years) were included. Four-point Dixon MRI scans of the right upper arm were performed at baseline and at the 12-, 18-, or 24-month follow-up. Elbow flexor FFs were determined from 5 central slices. Loss of hand-to-mouth movement was determined at study visits and by phone calls every 4 months. FFs were fitted to a sigmoidal curve by use of a mixed model with random slope to predict individual trajectories. The added predictive value of elbow flexor FF to age at loss of hand-to-mouth movement was calculated from a Cox model with the predicted FF as a time-varying covariate, yielding a hazard ratio. RESULTS Forty-eight MRIs of 20 patients with DMD were included. The hazard ratio of a percent-point increase in elbow flexor FF for the time to loss of hand-to-mouth movement was 1.12 (95% confidence interval 1.04-1.21; p = 0.002). This corresponded to a 3.13-fold increase in the instantaneous risk of loss of hand-to-mouth movement in patients with a 10-percent points higher elbow flexor FF at any age. DISCUSSION In this prospective study, elbow flexor FF predicted loss of hand-to-mouth movement independently of age. qMRI-measured elbow flexor FF can be used as a surrogate endpoint or stratification tool for clinical trials in nonambulant patients with DMD. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that qMRI FF of elbow flexor muscles in patients with DMD predicts loss of hand-to-mouth movement independently of age.
Collapse
Affiliation(s)
- Karin J Naarding
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Menno van der Holst
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erik W van Zwet
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Nienke M van de Velde
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Imelda J M de Groot
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan J G M Verschuuren
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hermien E Kan
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Erik H Niks
- From the Department of Neurology (K.J.N., N.M.v.d.V., J.J.G.M.V., E.H.N.), Duchenne Center Netherlands (K.J.N., M.v.d.H., N.M.v.d.V., I.J.M.d.G., J.J.G.M.V., H.E.K., E.H.N.)Department of Orthopedics, Rehabilitation and Physiotherapy (M.v.d.H.), and Department of Biomedical Data Sciences (E.W.v.Z.), Leiden University Medical Center; and Department of Rehabilitation (I.J.M.d.G.), Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
140
|
Salvadori L, Chiappalupi S, Arato I, Mancuso F, Calvitti M, Marchetti MC, Riuzzi F, Calafiore R, Luca G, Sorci G. Sertoli Cells Improve Myogenic Differentiation, Reduce Fibrogenic Markers, and Induce Utrophin Expression in Human DMD Myoblasts. Biomolecules 2021; 11:1504. [PMID: 34680138 PMCID: PMC8533898 DOI: 10.3390/biom11101504] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disease caused by mutations in DMD gene translating in lack of functional dystrophin and resulting in susceptibility of myofibers to rupture during contraction. Inflammation and fibrosis are critical hallmarks of DMD muscles, which undergo progressive degeneration leading to loss of independent ambulation in childhood and death by early adulthood. We reported that intraperitoneal injection of microencapsulated Sertoli cells (SeC) in dystrophic mice translates into recovery of muscle morphology and performance thanks to anti-inflammatory effects and induction of the dystrophin paralogue, utrophin at the muscle level, opening new avenues in the treatment of DMD. The aim of this study is to obtain information about the direct effects of SeC on myoblasts/myotubes, as a necessary step in view of a translational application of SeC-based approaches to DMD. We show that (i) SeC-derived factors stimulate cell proliferation in the early phase of differentiation in C2C12, and human healthy and DMD myoblasts; (ii) SeC delay the expression of differentiation markers in the early phase nevertheless stimulating terminal differentiation in DMD myoblasts; (iii) SeC restrain the fibrogenic potential of fibroblasts, and inhibit myoblast-myofibroblast transdifferentiation; and, (iv) SeC provide functional replacement of dystrophin in preformed DMD myotubes regardless of the mutation by inducing heregulin β1/ErbB2/ERK1/2-dependent utrophin expression. Altogether, these results show that SeC are endowed with promyogenic and antifibrotic effects on dystrophic myoblasts, further supporting their potential use in the treatment of DMD patients. Our data also suggest that SeC-based approaches might be useful in improving the early phase of muscle regeneration, during which myoblasts have to adequately proliferate to replace the damaged muscle mass.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Cell Proliferation/genetics
- Cell Transdifferentiation/genetics
- Disease Models, Animal
- Dystrophin/genetics
- Gene Expression Regulation/genetics
- Genetic Diseases, X-Linked/genetics
- Genetic Diseases, X-Linked/metabolism
- Genetic Diseases, X-Linked/pathology
- Humans
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Injections, Intraperitoneal
- MAP Kinase Signaling System/genetics
- Male
- Mice
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Myoblasts/metabolism
- Neuregulin-1/genetics
- Receptor, ErbB-2/genetics
- Regeneration/genetics
- Sertoli Cells/metabolism
- Sertoli Cells/pathology
- Utrophin/genetics
Collapse
Affiliation(s)
- Laura Salvadori
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy;
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (F.R.)
| | - Sara Chiappalupi
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (F.R.)
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
- Consorzio Interuniversitario Biotecnologie (CIB), 34127 Trieste, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
| | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
| | - Maria Cristina Marchetti
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
| | - Francesca Riuzzi
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (F.R.)
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
- Consorzio Interuniversitario Biotecnologie (CIB), 34127 Trieste, Italy
| | - Riccardo Calafiore
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
- Centro Biotecnologico Internazionale di Ricerca Traslazionale ad Indirizzo Endocrino, Metabolico ed Embrio-Riproduttivo (CIRTEMER), 06132 Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
- Centro Biotecnologico Internazionale di Ricerca Traslazionale ad Indirizzo Endocrino, Metabolico ed Embrio-Riproduttivo (CIRTEMER), 06132 Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale (CURGeF), 06132 Perugia, Italy
| | - Guglielmo Sorci
- Interuniversity Institute of Myology (IIM), 06132 Perugia, Italy; (S.C.); (F.R.)
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy; (I.A.); (F.M.); (M.C.); (M.C.M.); (R.C.)
- Consorzio Interuniversitario Biotecnologie (CIB), 34127 Trieste, Italy
- Centro Biotecnologico Internazionale di Ricerca Traslazionale ad Indirizzo Endocrino, Metabolico ed Embrio-Riproduttivo (CIRTEMER), 06132 Perugia, Italy
- Centro Universitario di Ricerca sulla Genomica Funzionale (CURGeF), 06132 Perugia, Italy
| |
Collapse
|
141
|
Elhussieny A, Nogami K, Sakai-Takemura F, Maruyama Y, Takemura N, Soliman WT, Takeda S, Miyagoe-Suzuki Y. Mesenchymal stem cells derived from human induced pluripotent stem cells improve the engraftment of myogenic cells by secreting urokinase-type plasminogen activator receptor (uPAR). Stem Cell Res Ther 2021; 12:532. [PMID: 34627382 PMCID: PMC8501581 DOI: 10.1186/s13287-021-02594-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe X-linked recessive disease caused by mutations in the dystrophin gene. Transplantation of myogenic stem cells holds great promise for treating muscular dystrophies. However, poor engraftment of myogenic stem cells limits the therapeutic effects of cell therapy. Mesenchymal stem cells (MSCs) have been reported to secrete soluble factors necessary for skeletal muscle growth and regeneration. Methods We induced MSC-like cells (iMSCs) from induced pluripotent stem cells (iPSCs) and examined the effects of iMSCs on the proliferation and differentiation of human myogenic cells and on the engraftment of human myogenic cells in the tibialis anterior (TA) muscle of NSG-mdx4Cv mice, an immunodeficient dystrophin-deficient DMD model. We also examined the cytokines secreted by iMSCs and tested their effects on the engraftment of human myogenic cells. Results iMSCs promoted the proliferation and differentiation of human myogenic cells to the same extent as bone marrow-derived (BM)-MSCs in coculture experiments. In cell transplantation experiments, iMSCs significantly improved the engraftment of human myogenic cells injected into the TA muscle of NSG-mdx4Cv mice. Cytokine array analysis revealed that iMSCs produced insulin-like growth factor-binding protein 2 (IGFBP2), urokinase-type plasminogen activator receptor (uPAR), and brain-derived neurotrophic factor (BDNF) at higher levels than did BM-MSCs. We further found that uPAR stimulates the migration of human myogenic cells in vitro and promotes their engraftment into the TA muscles of immunodeficient NOD/Scid mice. Conclusions Our results indicate that iMSCs are a new tool to improve the engraftment of myogenic progenitors in dystrophic muscle. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02594-1.
Collapse
Affiliation(s)
- Ahmed Elhussieny
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Neurology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Ken'ichiro Nogami
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Fusako Sakai-Takemura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Yusuke Maruyama
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.,Department of Gene Regulation, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Natsumi Takemura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Wael Talaat Soliman
- Department of Neurology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Yuko Miyagoe-Suzuki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
142
|
Wang L, Li H, Li Y, Xu M, Lin J, Liao Z, Pei Z, Zhang C. Generation and characterization of an induced pluripotent stem cell line (ZSYYDNi001-A) from a patient with Duchenne muscular dystrophy carrying exon 51 deletion in the DMD gene. Stem Cell Res 2021; 56:102553. [PMID: 34619646 DOI: 10.1016/j.scr.2021.102553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/05/2021] [Accepted: 09/26/2021] [Indexed: 11/26/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a common hereditary neuromuscular disease characterized by progressive muscle wasting and weakness. DMD is caused by mutations in the DMD gene, resulting in the dysfunction of dystrophin. We generated an induced pluripotent stem cell (iPSC) from a patient with DMD carrying exon 51 deletion in the DMD gene. This iPSC line can serve as a model for studying the pathogenesis and therapeutics of DMD.
Collapse
Affiliation(s)
- Liang Wang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Huan Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Yaqin Li
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China
| | - Min Xu
- Department of Dermatology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 510260, China
| | - Jinfu Lin
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Ziyu Liao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China.
| | - Cheng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou 510080, China.
| |
Collapse
|
143
|
Trucco F, Ridout D, Domingos J, Maresh K, Chesshyre M, Munot P, Sarkozy A, Robb S, Quinlivan R, Riley M, Wallis C, Chan E, Abel F, De Lucia S, Hogrel JY, Niks EH, de Groot I, Servais L, Straub V, Ricotti V, Manzur A, Muntoni F. Genotype-related respiratory progression in Duchenne muscular dystrophy-A multicenter international study. Muscle Nerve 2021; 65:67-74. [PMID: 34606104 DOI: 10.1002/mus.27427] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 01/26/2023]
Abstract
INTRODUCTION/AIMS Mutations amenable to skipping of specific exons have been associated with different motor progression in Duchenne muscular dystrophy (DMD). Less is known about their association with long-term respiratory function. In this study we investigated the features of respiratory progression in four DMD genotypes relevant in ongoing exon-skipping therapeutic strategies. METHODS This was a retrospective longitudinal study including DMD children followed by the UK NorthStar Network and international AFM Network centers (May 2003 to October 2020). We included boys amenable to skip exons 44, 45, 51, or 53, who were older than 5 years of age and ambulant at first recorded visit. Subjects who were corticosteroid-naive or enrolled in interventional clinical trials were excluded. The progression of respiratory function (absolute forced vital capacity [FVC] and calculated as percent predicted [FVC%]) was compared across the four subgroups (skip44, skip45, skip51, skip53). RESULTS We included 142 boys in the study. Mean (standard deviation) age at first visit was 8.6 (2.5) years. Median follow-up was 3 (range, 0.3-8.3) years. In skip45 and skip51, FVC% declined linearly from the first recorded visit. From the age of 9 years, FVC% declined linearly in all genotypes. Skip44 had the slowest (2.7%/year) and skip51 the fastest (5.9%/year) annual FVC% decline. The absolute FVC increased progressively in skip44, skip45, and skip51. In skip53, FVC started declining from 14 years of age. DISCUSSION The progression of respiratory dysfunction follows different patterns for specific genotype categories. This information is valuable for prognosis and for the evaluation of exon-skipping therapies.
Collapse
Affiliation(s)
- Federica Trucco
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK.,Department Paediatric Neuroscience, Guy's and St Thomas NHS Trust and Department Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Deborah Ridout
- Population, Policy and Practice Research and Teaching Department, University College London Great Ormond Street Institute of Child Health, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, UK
| | - Joana Domingos
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Kate Maresh
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Mary Chesshyre
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Pinki Munot
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Stephanie Robb
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Rosaline Quinlivan
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK.,MRC Centre for Neuromuscular Disease, National Hospital for Neurology and Neurosurgery, London, UK
| | - Mollie Riley
- Lung Function Laboratory, Great Ormond Street Hospital, London, UK
| | - Colin Wallis
- Department of Respiratory Medicine, Great Ormond Street Hospital, London, UK
| | - Elaine Chan
- Department of Respiratory Medicine, Great Ormond Street Hospital, London, UK
| | - Francois Abel
- Department of Respiratory Medicine, Great Ormond Street Hospital, London, UK
| | | | | | - Erik H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Imelda de Groot
- Department of Rehabilitation, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Laurent Servais
- Centre de Référence Des Maladies Neuromusculaires, CHU de Liège, Liège, Belgium.,Department of Paediatrics, MDUK Neuromuscular Center, University of Oxford, Oxford, UK
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Valeria Ricotti
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, UK
| | - Adnan Manzur
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, University College London Great Ormond Street Institute of Child Health and Great Ormond Street Hospital, London, UK.,NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, UK
| | | |
Collapse
|
144
|
Landfeldt E, Iff J, Henricson E. Rasch Analysis of the Pediatric Quality of Life Inventory 4.0 Generic Core Scales Administered to Patients With Duchenne Muscular Dystrophy. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2021; 24:1490-1498. [PMID: 34593173 PMCID: PMC9132346 DOI: 10.1016/j.jval.2021.05.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 04/27/2021] [Accepted: 05/20/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVES The objective of this study was to examine the psychometric properties of the Pediatric Quality of Life Inventory 4.0 Generic Core Scales (PedsQL 4.0 GCS) in Duchenne muscular dystrophy (DMD), a rare, severely debilitating, and ultimately fatal neuromuscular disease. METHODS Patients with DMD were recruited from 20 centers across 9 countries as part of the Cooperative International Neuromuscular Research Group Duchenne Natural History Study (NCT00468832). The psychometric properties of the PedsQL 4.0 GCS were examined using Rasch analysis. RESULTS In total, 329 patients with DMD (mean age 9 years, range 3-18 years, 75% ambulatory) completed the PedsQL 4.0 GCS. The most difficult instrument items, expressing the greatest loss in health-related quality of life, were those associated with emotional well-being (eg, being teased by other children, feeling sad, and not making friends), as opposed to somatic disability (eg, lifting heavy objects, participating in sports, and running). The mean item and person fit residuals were estimated at 0.301 (SD: 1.385) and -0.255 (1.504), respectively. In total, 87% (20 of 23) of items displayed disordered thresholds, and many exhibited nontrivial dependency. The overall item-trait interaction χ2 value was 178 (115 degrees of freedom, P<.001). Our analysis also revealed significant issues with differential item functioning, and by investigating residual principal component loadings, the PedsQL 4.0 GCS total score was found to be multidimensional. CONCLUSIONS The PedsQL 4.0 GCS records information clinically relevant to patients with DMD, but the total scale score may not be fit for purpose as a measure health-related quality of life in this disease population.
Collapse
Affiliation(s)
- Erik Landfeldt
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; ICON plc, Stockholm, Sweden.
| | - Joel Iff
- Sarepta Therapeutics Inc, Cambridge, MA, USA
| | - Erik Henricson
- Department of Physical Medicine and Rehabilitation, University of California Davis School of Medicine, Sacramento, CA, USA
| |
Collapse
|
145
|
An Overview of Physical Exercise and Antioxidant Supplementation Influences on Skeletal Muscle Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10101528. [PMID: 34679663 PMCID: PMC8532825 DOI: 10.3390/antiox10101528] [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: 07/24/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
One of the essential injuries caused by moderate to high-intensity and short-duration physical activities is the overproduction of reactive oxygen species (ROS), damaging various body tissues such as skeletal muscle (SM). However, ROS is easily controlled by antioxidant defense systems during low to moderate intensity and long-term exercises. In stressful situations, antioxidant supplements are recommended to prevent ROS damage. We examined the response of SM to ROS generation during exercise using an antioxidant supplement treatment strategy in this study. The findings of this review research are paradoxical due to variances in antioxidant supplements dose and duration, intensity, length, frequency, types of exercise activities, and, in general, the lack of a regular exercise and nutrition strategy. As such, further research in this area is still being felt.
Collapse
|
146
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [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: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
147
|
Duan D. A Cautiously Optimistic Outlook of a Designer Therapy for 1% of Duchenne Muscular Dystrophy Patients. Hum Gene Ther 2021; 32:872-874. [PMID: 34554886 DOI: 10.1089/hum.2021.29179.ddu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dongsheng Duan
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, Missouri, USA.,Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA.,Department of Biomedical, Biological and Chemical Engineering, College of Engineering, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
148
|
Sun Z, Xu D, Zhao L, Li X, Li S, Huang X, Li C, Sun L, Liu B, Jiang Z, Zhang L. A new therapeutic effect of fenofibrate in Duchenne muscular dystrophy: The promotion of myostatin degradation. Br J Pharmacol 2021; 179:1237-1250. [PMID: 34553378 DOI: 10.1111/bph.15678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/14/2021] [Accepted: 08/19/2021] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Duchenne muscular dystrophy (DMD) is a degenerative muscle disease with no effective drug treatment. This study investigated the positive effects of fenofibrate on dystrophic muscles. EXPERIMENTAL APPROACH Myostatin expression in serum and muscle tissue of DMD patients and mdx mice were tested. Primary myoblasts isolated from mdx mice were challenged with an inflammatory stimulus and treated with fenofibrate. In animal experiments, 6-week-old male mdx mice were treated with fenofibrate (100 mg/kg) administered orally once per day for 6 weeks. Tests of muscle function plus histology and biochemical analyses of serum were conducted to evaluate the effects of fenofibrate. The expressions of myostatin, MuRF1, and atrogin-1 in skeletal muscle were evaluated by Western blotting and real-time PCR. Total and oxidative myosin heavy chain (MHC) were assessed via immunofluorescence. KEY RESULTS Increased expression of myostatin protein was found in dystrophic muscle of DMD patients and mdx mice. Fenofibrate enhanced myofibre differentiation by downregulating the expression of myostatin protein but not mRNA in primary myoblasts of mdx mice. Fenofibrate significantly improved muscle function while ameliorating muscle damage in mdx mice. These benefits are accompanied by an anti-inflammatory effect. Fenofibrate treatment returned myofibre function by inhibiting the expressions of myostatin, MuRF1, and atrogin-1 protein in the gastrocnemius muscle and diaphragm, while leaving the mRNA level of myostatin unaffected. CONCLUSIONS AND IMPLICATIONS Fenofibrate substantially slows muscle dystrophy by promoting the degradation of myostatin protein, which may indicate a new therapeutic focus for DMD patients.
Collapse
Affiliation(s)
- Zeren Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Dengqiu Xu
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lei Zhao
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Xihua Li
- Department of Neurology, Children's Hospital of Fudan University, Shanghai, China
| | - Sijia Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Xiaofei Huang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Chunjie Li
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Lixin Sun
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China
| | - Bing Liu
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhenzhou Jiang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Luyong Zhang
- Jiangsu Key Laboratory of Drug Screening, Key Laboratory of Drug Quality Control and Pharmacovigilance, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, China Pharmaceutical University, Nanjing, China.,Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
149
|
TERAMOTO N, IKEDA M, SUGIHARA H, SHIGA T, MATSUWAKI T, NISHIHARA M, UCHIDA K, YAMANOUCHI K. Loss of p16/Ink4a drives high frequency of rhabdomyosarcoma in a rat model of Duchenne muscular dystrophy. J Vet Med Sci 2021; 83:1416-1424. [PMID: 34334511 PMCID: PMC8498826 DOI: 10.1292/jvms.21-0243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022] Open
Abstract
Rhabdomyosarcoma (RMS) is an aggressive type of soft tissue sarcoma, and pleomorphic RMS is a rare subtype of RMS found in adult. p16 is a tumor suppressor which inhibits cell cycle. In human RMS, p16 gene is frequently deleted, but p16-null mice do not develop RMS. We reported that genetic ablation of p16 by the crossbreeding of p16 knock-out rats (p16-KO rats) improved the dystrophic phenotype of a rat model of Duchenne muscular dystrophy (Dmd-KO rats). However, p16/Dmd double knock-out rats (dKO rats) unexpectedly developed sarcoma. In the present study, we raised p16-KO, Dmd-KO, and dKO rats until 11 months of age. Twelve out of 22 dKO rats developed pleomorphic RMS after 9 months of age, while none of p16-KO rats and Dmd-KO rats developed tumor. The neoplasms were connected to skeletal muscle tissue with indistinct borders and characterized by diffuse proliferation of pleomorphic cells which had eosinophilic cytoplasm and atypical nuclei with anisokaryosis. For almost all cases, the tumor cells immunohistochemically expressed myogenic markers including desmin, MyoD, and myogenin. The single cell cloning from tumor primary cells gained 20 individual Pax7-negative MyoD-positive RMS cell clones. Our results demonstrated that double knock-out of p16 and dystrophin in rats leads to the development of pleomorphic RMS, providing an animal model that may be useful to study the developmental mechanism of pleomorphic RMS.
Collapse
Affiliation(s)
- Naomi TERAMOTO
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Masanari IKEDA
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hidetoshi SUGIHARA
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Takanori SHIGA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Takashi MATSUWAKI
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Masugi NISHIHARA
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Kazuyuki UCHIDA
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Keitaro YAMANOUCHI
- Laboratory of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| |
Collapse
|
150
|
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked, muscle wasting disease that affects 1 in 5000 males. Affected individuals become wheelchair bound by the age of twelve and eventually die in their third decade due to respiratory and cardiac complications. The disease is caused by mutations in the DMD gene that codes for dystrophin. Dystrophin is a structural protein that maintains the integrity of muscle fibres and protects them from contraction-induced damage. The absence of dystrophin compromises the stability and function of the muscle fibres, eventually leading to muscle degeneration. So far, there is no effective treatment for deteriorating muscle function in DMD patients. A promising approach for treating this life-threatening disease is gene transfer to restore dystrophin expression using a safe, non-pathogenic viral vector called adeno-associated viral (AAV) vector. Whilst microdystrophin gene transfer using AAV vectors shows extremely impressive therapeutic success so far in large animal models of DMD, translating this advanced therapy medicinal product from bench to bedside still offers scope for many optimization steps. In this paper, the authors review the current progress of AAV-microdystrophin gene therapy for DMD and other treatment strategies that may apply to a subset of DMD patients depending on the mutations they carry.
Collapse
Affiliation(s)
- Nertiyan Elangkovan
- Centres for Gene & Cell Therapy and Biomedical Sciences, Department of Biological Sciences, School of Life & Environmental Sciences, Royal Holloway - University of London, Surrey, TW20 0EX, UK
| | - George Dickson
- Centres for Gene & Cell Therapy and Biomedical Sciences, Department of Biological Sciences, School of Life & Environmental Sciences, Royal Holloway - University of London, Surrey, TW20 0EX, UK
| |
Collapse
|