1
|
Xie Y, Luo J, Zhong J, Lan D. Clinical Diagnosis and Genetic Analysis of Children With Muscular Dystrophies. Clin Pediatr (Phila) 2024:99228241272029. [PMID: 39198981 DOI: 10.1177/00099228241272029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/01/2024]
Abstract
More than 90% suspected muscular dystrophy (MD) can be confirmed with multiplex ligation-dependent probe amplification (MLPA) combined with targeted panel, although there are a few that cannot be identified. A total of 312 suspected MD patients were enrolled into the study. The MLPA combined with a targeted myopathy panel were performed. Patients with negative results were subjected to whole exome sequencing (WES), whole genome sequencing (WGS), and/or RNA sequencing (RNA-seq). A total of 275 cases were diagnosed as Duchenne/Becker muscular dystrophy (DMD/BMD) and 20 cases were other types of myopathy or nonmuscular diseases. Six female DMD/BMD patients suffered from varying degrees of typical DMD-like symptoms and 2 others were suspected to be gonadal mosaicism. The systematic application of WES, WGS, and/or RNA-seq highlighted the need for the detection of variants missed by the current standard diagnostic procedures. The identification of female patients and mosaic carriers was crucial to predict the risk of recurrence and allow for optimal genetic counseling.
Collapse
Affiliation(s)
- Yanshu Xie
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingsi Luo
- Genetic and Metabolic Central Laboratory, Guangxi Birth Defects Research and Prevention Institute, The Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Guangxi Key Laboratory of Precision Medicine for Genetic Diseases, The Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jingzi Zhong
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dan Lan
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Bever A, Audhya I, Szabo SM, Mickle A, Feeny D, Malone D, Neumann P, Iannaccone S, Gooch K. "You Take This Day by Day, Come What May": A Qualitative Study of the Psychosocial Impacts of Living with Duchenne Muscular Dystrophy. Adv Ther 2024; 41:2460-2476. [PMID: 38709395 PMCID: PMC11133021 DOI: 10.1007/s12325-024-02867-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION Studies have reported health-related quality-of-life impacts of Duchenne muscular dystrophy (DMD); however, further research is needed to understand how those with DMD experience their condition and how psychosocial impacts evolve over time in response to disease progression. This qualitative study explores the social and emotional implications of key transitions, challenges and adaptations throughout the disease course from the perspective of patients and family caregivers. METHODS Semi-structured interviews were conducted with men and boys with DMD, and/or their caregivers, in the USA. Thematic analysis was used to examine patterns in data collected across the interviews. RESULTS Nineteen participants were included. Three major themes were identified: (1) barriers to participation are multifaceted; (2) an emotional journey shaped by 'inevitable progression;' (3) family provides critical tangible and emotional support. This study illustrates that psychosocial impacts of DMD are shaped by knowledge of the condition's natural history alongside other factors including the extent of social barriers, personal growth and adaptation, and family support. CONCLUSIONS Findings provide insight into the strength and resilience with which individuals and their families respond to daily challenges and major clinical milestones and highlight the relative importance of loss of upper limb function as a transition in DMD affecting health-related quality-of-life.
Collapse
Affiliation(s)
- Andrea Bever
- Broadstreet HEOR, 201-343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - Ivana Audhya
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA, USA
| | - Shelagh M Szabo
- Broadstreet HEOR, 201-343 Railway St, Vancouver, BC, V6A 1A4, Canada.
| | - Alexis Mickle
- Broadstreet HEOR, 201-343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - David Feeny
- McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4L8, Canada
| | - Daniel Malone
- The University of Utah, 201 Presidents Circle, Salt Lake City, UT, 84112, USA
| | - Peter Neumann
- Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA
| | - Susan Iannaccone
- The University of Texas Southwestern, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Katherine Gooch
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA, USA
| |
Collapse
|
3
|
Johnston KM, Audhya IF, Dunne J, Feeny D, Neumann P, Malone DC, Szabo SM, Gooch KL. Comparing Preferences for Disease Profiles: A Discrete Choice Experiment from a US Societal Perspective. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2024; 22:343-352. [PMID: 38253973 PMCID: PMC11021240 DOI: 10.1007/s40258-023-00869-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/24/2024]
Abstract
OBJECTIVES There is increasing interest in expanding the elements of value to be considered when making health policy decisions. To help inform value frameworks, this study quantified preferences for disease attributes in a general public sample and examined which combination of attributes (disease profiles) are considered most important for research and treatment. METHODS A discrete choice experiment (DCE) was conducted in a US general population sample, recruited through online consumer panels. Respondents were asked to select one of a set of health conditions they believed to be most important, characterized by attributes defined by a previous qualitative study: onset age; cause of disease; life expectancy; caregiver requirement; symptom burden (characterized by the Health Utilities Index with varying levels of ambulation independence, dexterity limitations, and degree of pain and discomfort); and disease prevalence. A fractional factorial DCE design was implemented using R, and 60 choice sets were generated (separated into blocks of 10 per participant). Data were analyzed using a mixed-logit regression model, and results used to assess the likelihood of preferring disease profiles. Based on individual attribute preferences, overall preferences for disease profiles, including a profile aligned with Duchenne muscular dystrophy (DMD), were compared. RESULTS Fifty-two percent of respondents (n = 537) were female, and 70.6% were aged 18-54 years. Attributes considered most important were those related to life expectancy (odds ratio [OR], 95% confidence interval [CI] 1.88 [1.56-2.27] for a 50% reduction in remaining life expectancy vs no impact), and symptom burden (OR [95% CI] 1.84 [1.47-2.31] for severe vs mild burden). Greater importance was also found for pediatric onset, caregiver requirement, and diseases affecting more people. As an example of disease profile preferences, a DMD-like pediatric inherited disease with 50% reduction in life expectancy, extensive caregiver requirement, severe symptom burden, and 1:5000 prevalence had 2.37-fold higher odds of being selected as important versus an equivalent disease with adult onset and no life expectancy reduction. CONCLUSIONS Of disease attributes included in this DCE, respondents valued higher prevalence of disease, life expectancy and symptom burden as most important for prioritizing research and treatment. Based on expressed attribute preferences, a case study of an inherited pediatric disease involving substantial reductions to length and quality of life and requiring caregiver support has relatively high odds of being identified as important compared to diseases reflecting differing attribute profiles. These findings can help inform expansions of value frameworks by identifying important attributes from the societal perspective.
Collapse
Affiliation(s)
| | - Ivana F Audhya
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA, USA
| | - Jessica Dunne
- Broadstreet HEOR, 201-343 Railway St., Vancouver, BC, V6A 1A4, Canada
| | | | | | | | - Shelagh M Szabo
- Broadstreet HEOR, 201-343 Railway St., Vancouver, BC, V6A 1A4, Canada
| | | |
Collapse
|
4
|
Clavero-Adell M, Ayerza-Casas A, Palanca-Arias D, López-Ramón M, Jiménez-Montañés L, Olmos S. Early assessment of cardiomyopathy in Duchenne patients by means of longitudinal strain echocardiography. Cardiol Young 2024; 34:151-156. [PMID: 37272541 DOI: 10.1017/s104795112300149x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
INTRODUCTION The diagnosis of Duchenne-linked cardiomyopathy may be challenging. Conventional echocardiographic measurements typically show deterioration beyond the second decade. Global longitudinal strain has been proposed as an earlier marker than left ventricular ejection fraction. MATERIAL AND METHODS A prospective, observational, cross-sectional, case-control study was carried out. Both Duchenne patients and control subjects underwent transthoracic echocardiogram in order to assess left ventricle function. Bayesian linear regression was the main tool for inference. Age effects were parameterised by means of a spline function because of its simplicity to characterise continuous variables and smooth contributions. The posterior distribution of the marginal age effects was used to assess the earliest age of deterioration of each marker. RESULTS Sixteen Duchenne patients and twenty-two healthy male subjects were enrolled. On overage, cardiac function measures were found for ejection fraction and longitudinal strain among different groups. Age effects on global longitudinal strain are more reliably found at ages of 6 and above, while ejection fraction starts to deteriorate at an older age. Progressive left ventricular dysfunction in Duchenne patients is one of the key issues and starts at an early age with subtle symptoms. CONCLUSION This cross-sectional study provides supporting evidence that global longitudinal strain is an earlier marker of disease progression than ejection fraction in Duchenne patients.
Collapse
Affiliation(s)
- Marcos Clavero-Adell
- Pediatric Cardiology Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Dislipemias Primarias, IIS Aragón, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Medicine, Universidad de Zaragoza, Spain
| | - Ariadna Ayerza-Casas
- Pediatric Cardiology Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Faculty of Medicine, Universidad de Zaragoza, Spain
| | - Daniel Palanca-Arias
- Pediatric Cardiology Unit, University Hospital Miguel Servet, Zaragoza, Spain
- Faculty of Medicine, Universidad de Zaragoza, Spain
| | - Marta López-Ramón
- Pediatric Cardiology Unit, University Hospital Miguel Servet, Zaragoza, Spain
| | | | - Salvador Olmos
- Dislipemias Primarias, IIS Aragón, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBER-CV), Instituto de Salud Carlos III, Madrid, Spain
- Aragon Institute of Engineering Research (I3A), Universidad de Zaragoza, Spain
| |
Collapse
|
5
|
Mashima R, Takada S, Miyamoto Y. RNA-Based Therapeutic Technology. Int J Mol Sci 2023; 24:15230. [PMID: 37894911 PMCID: PMC10607345 DOI: 10.3390/ijms242015230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
RNA-based therapy has been an expanding area of clinical research since the COVID-19 outbreak. Often, its comparison has been made to DNA-based gene therapy, such as adeno-associated virus- and lentivirus-mediated therapy. These DNA-based therapies show persistent expression, with maximized therapeutic efficacy. However, accumulating data indicate that proper control of gene expression is occasionally required. For example, in cancer immunotherapy, cytokine response syndrome is detrimental for host animals, while excess activation of the immune system induces supraphysiological cytokines. RNA-based therapy seems to be a rather mild therapy, and it has room to fit unmet medical needs, whereas current DNA-based therapy has unclear issues. This review focused on RNA-based therapy for cancer immunotherapy, hematopoietic disorders, and inherited disorders, which have received attention for possible clinical applications.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yoshitaka Miyamoto
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
6
|
Audhya I, Rogula B, Szabo SM, Feeny D, Bolatova T, Gooch K. Exploring the relationship between North Star Ambulatory Assessment and Health Utilities Index scores in Duchenne muscular dystrophy. Health Qual Life Outcomes 2023; 21:76. [PMID: 37468890 PMCID: PMC10355009 DOI: 10.1186/s12955-023-02160-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND The North Star Ambulatory Assessment (NSAA) documents motor performance in ambulatory individuals with Duchenne muscular dystrophy (DMD). Health Utilities Index (HUI) scores, reflecting preferences for health-related quality-of-life (HRQoL) implications of health states, are commonly estimated within trials. This study sought to characterize the relationship between the NSAA score and utility in DMD. METHODS Family members serving as proxy respondents for placebo-treated ambulatory individuals with DMD (NCT01254019; BioMarin Pharmaceuticals Inc) completed the HUI and the NSAA (score range, 0-34). Mean change over time on these measures was estimated, and the correlation between changes in NSAA score and a) HUI utility; b) HUI3 ambulation and HUI2 mobility attribute scores, over 48 weeks was calculated. RESULTS Baseline mean (range) age was 8.0 years (5-16; n = 61) and mean (standard deviation [SD]) scores were 0.87 (0.13; HUI2), 0.82 (0.19; HUI3), and 21.0 (8.1; NSAA). Mean (SD) change over 48 weeks was -0.05 (0.14; HUI2), -0.06 (0.19; HUI3), and -2.9 (4.7; NSAA). Weak positive correlations were observed between baseline NSAA score and HUI utility (HUI2: r = 0.29; HUI3: r = 0.17) and for change over 48 weeks (HUI2: r = 0.16; HUI3: r = 0.15). Stronger correlations were observed between change in NSAA score and the HUI3 ambulation (r = 0.41) and HUI2 mobility (r = 0.41) attributes. CONCLUSIONS Among ambulatory individuals with DMD, NSAA score is weakly correlated with HUI utility, suggesting that motor performance alone does not fully explain HRQoL. Stronger relationships were observed between HUI ambulation and mobility attributes, and NSAA. Although unidimensional measures like the NSAA are informative for documenting disease-specific health impacts, they may not correlate well with measures of overall health status; requiring use in conjunction with other patient-reported and preference-based outcomes.
Collapse
Affiliation(s)
| | - Basia Rogula
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada
| | - Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada.
| | - David Feeny
- McMaster University and Health Utilities Inc, Hamilton, ON, Canada
| | - Talshyn Bolatova
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A6, Canada
| | | |
Collapse
|
7
|
Czifrus E, Berlau DJ. Viltolarsen: a treatment option for Duchenne muscular dystrophy patients who are amenable to exon 53 skipping therapy. Expert Rev Neurother 2023; 23:853-858. [PMID: 37572081 DOI: 10.1080/14737175.2023.2246658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a progressive genetic disease leading to muscular weakness. DMD is caused by mutations of the dystrophin gene on the X chromosome that is responsible for production of dystrophin protein. Dystrophin contributes to structural support in muscle cells and mutations result in dystrophin protein deficiency which causes muscle damage and the associated clinical presentation. Exon skipping medications, including the exon 53 targeting viltolarsen, are the first agents with the ability to partially restore dystrophin protein. AREAS COVERED Herein, the authors profile viltolarsen for the DMD patients who are amenable to exon 53 skipping therapy and provide their expert perspectives on this subject. EXPERT OPINION Current findings suggest that viltolarsen could play a role in the current and possible future treatment of DMD. Viltolarsen seems to be safe and restores dystrophin protein to around 6% of the normal level. Due to orphan drug status, after the completion of the phase 2 clinical trial, viltolarsen was granted accelerated approval in Japan and in the US. A phase 3 trial is currently in progress and needs to earn full approval. Although a multidisciplinary approach continues to be critical, the addition of exon skipping agents like viltolarsen may improve the quality of patients' lives. However, data on the long-term safety and efficacy of this medication are not yet available due to its recent accelerated approval.
Collapse
Affiliation(s)
- Eszter Czifrus
- Semmelweis University Faculty of Medicine, Budapest, Hungary
| | - Daniel J Berlau
- Department of Pharmaceutical Sciences, Regis University School of Pharmacy, Denver, CO, USA
| |
Collapse
|
8
|
Tang F, Xiao Y, Zhou C, Zhang H, Wang J, Zeng Y. NGS-based targeted sequencing identified six novel variants in patients with Duchenne/Becker muscular dystrophy from southwestern China. BMC Med Genomics 2023; 16:121. [PMID: 37254189 DOI: 10.1186/s12920-023-01556-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND At present, Multiplex ligation-dependent probe amplification (MLPA) and exome sequencing are common gene detection methods in patients with Duchenne muscular dystrophy or Becker muscular dystrophy (DMD/BMD), but they can not cover the whole-genome sequence of the DMD gene. In this study, the whole genome capture of the DMD gene and next-generation sequencing (NGS) technology were used to detect the patients with DMD/BMD in Southwest China, to clarify the application value of this technology and further study the gene variant spectrum. METHODS From 2017 to 2020, 51 unrelated patients with DMD/BMD in southwestern China were clinically diagnosed at West China Second University Hospital of Sichuan University (Chengdu, China). The whole-genome of the DMD gene was captured from the peripheral blood of all patients, and next-generation sequencing was performed. Large copy number variants (CNVs) in the exon regions of the DMD gene were verified through MLPA, and small variations (such as single nucleotide variation and < 50 bp fragment insertions/deletions) were validated using Sanger sequencing. RESULTS Among the 51 patients, 49 (96.1% [49/51]) had pathogenic or likely pathogenic variants in the DMD gene. Among the 49 positive samples, 17 patients (34.7% [17/49]) had CNVs in the exon regions and 32 patients (65.3% [32/49]) had small variations. A total of six novel variants were identified: c.10916_10917del, c.1790T>A, c.1842del, c.5015del, c.5791_5792insCA, and exons 38-50 duplication. CONCLUSIONS Pathogenic or likely pathogenic variants of the DMD gene were detected in 49 patients (96.1% [49/51]), of which 6 variants (12.2% [6/49]) had not been previously reported. This study confirmed the value of NGS-based targeted sequencing for the DMD gene expanding the spectrum of variants in DMD, which may provide effective genetic counseling and prenatal diagnosis for families.
Collapse
Affiliation(s)
- Feng Tang
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuanyuan Xiao
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Cong Zhou
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Haixia Zhang
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jing Wang
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yang Zeng
- Department of Medical Genetics, West China Second University Hospital of Sichuan University, Chengdu, People's Republic of China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
9
|
Clarke LA, Amaral MD. What Can RNA-Based Therapy Do for Monogenic Diseases? Pharmaceutics 2023; 15:pharmaceutics15010260. [PMID: 36678889 PMCID: PMC9863139 DOI: 10.3390/pharmaceutics15010260] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
The use of RNA-based approaches to treat monogenic diseases (i.e., hereditary disorders caused by mutations in single genes) has been developed on different fronts. One approach uses small antisense oligonucleotides (ASOs) to modulate RNA processing at various stages; namely, to enhance correct splicing, to stimulate exon skipping (to exclude premature termination codon variants), to avoid undesired messenger RNA (mRNA) transcript degradation via the nonsense-mediated decay (NMD) pathway, or to induce mRNA degradation where they encode toxic proteins (e.g., in dominant diseases). Another approach consists in administering mRNA, which, like gene therapy, is a mutation-agnostic approach with potential application to any recessive monogenic disease. This is simpler than gene therapy because instead of requiring targeting of the nucleus, the mRNA only needs to be delivered to the cytoplasm. Although very promising (as demonstrated by COVID-19 vaccines), these approaches still have potential for optimisation, namely regarding delivery efficiency, adverse drug reactions and toxicity.
Collapse
|
10
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
11
|
Gomez Limia C, Baird M, Schwartz M, Saxena S, Meyer K, Wein N. Emerging Perspectives on Gene Therapy Delivery for Neurodegenerative and Neuromuscular Disorders. J Pers Med 2022; 12:1979. [PMID: 36556200 PMCID: PMC9788053 DOI: 10.3390/jpm12121979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 12/05/2022] Open
Abstract
Neurodegenerative disorders (NDDs), such as Alzheimer's disease (AD) and Parkinson's Disease (PD), are a group of heterogeneous diseases that mainly affect central nervous system (CNS) functions. A subset of NDDs exhibit CNS dysfunction and muscle degeneration, as observed in Gangliosidosis 1 (GM1) and late stages of PD. Neuromuscular disorders (NMDs) are a group of diseases in which patients show primary progressive muscle weaknesses, including Duchenne Muscular Dystrophy (DMD), Pompe disease, and Spinal Muscular Atrophy (SMA). NDDs and NMDs typically have a genetic component, which affects the physiological functioning of critical cellular processes, leading to pathogenesis. Currently, there is no cure or efficient treatment for most of these diseases. More than 200 clinical trials have been completed or are currently underway in order to establish safety, tolerability, and efficacy of promising gene therapy approaches. Thus, gene therapy-based therapeutics, including viral or non-viral delivery, are very appealing for the treatment of NDDs and NMDs. In particular, adeno-associated viral vectors (AAV) are an attractive option for gene therapy for NDDs and NMDs. However, limitations have been identified after systemic delivery, including the suboptimal capacity of these therapies to traverse the blood-brain barrier (BBB), degradation of the particles during the delivery, high reactivity of the patient's immune system during the treatment, and the potential need for redosing. To circumvent these limitations, several preclinical and clinical studies have suggested intrathecal (IT) delivery to target the CNS and peripheral organs via cerebrospinal fluid (CSF). CSF administration can vastly improve the delivery of small molecules and drugs to the brain and spinal cord as compared to systemic delivery. Here, we review AAV biology and vector design elements, different therapeutic routes of administration, and highlight CSF delivery as an attractive route of administration. We discuss the different aspects of neuromuscular and neurodegenerative diseases, such as pathogenesis, the landscape of mutations, and the biological processes associated with the disease. We also describe the hallmarks of NDDs and NMDs as well as discuss current therapeutic approaches and clinical progress in viral and non-viral gene therapy and enzyme replacement strategies for those diseases.
Collapse
Affiliation(s)
- Cintia Gomez Limia
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Megan Baird
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Maura Schwartz
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
| | - Smita Saxena
- Department of Neurology, Inselspital, 3010 Bern, Switzerland
| | - Kathrin Meyer
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| | - Nicolas Wein
- Center for Gene Therapy, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43205, USA
| |
Collapse
|
12
|
Wu X, Dong N, Yu L, Liu M, Jiang J, Tang T, Zhao H, Fang Q. Identification of immune-related features involved in Duchenne muscular dystrophy: A bidirectional transcriptome and proteome-driven analysis. Front Immunol 2022; 13:1017423. [PMID: 36483550 PMCID: PMC9724784 DOI: 10.3389/fimmu.2022.1017423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/02/2022] [Indexed: 11/23/2022] Open
Abstract
Background We aimed to investigate the biological mechanism and feature genes of Duchenne muscular dystrophy (DMD) by multi-omics and experimental verification strategy. Methods We integrated the transcriptomic and proteomic methods to find the differentially expressed mRNAs (DEMs) and proteins (DEPs) between DMD and Control groups. Weighted gene co-expression network analysis (WGCNA) was then used to identify modules of highly correlated genes and hub genes. In the following steps, the immune and stromal cells infiltrations were accomplished by xCELL algorithm. Furthermore, TF and miRNA prediction were performed with Networkanalyst. ELISA, western blot and external datasets were performed to verify the key proteins/mRNAs in DMD patient and mouse. Finally, a nomogram model was established based on the potential biomarkers. Results 4515 DEMs and 56 DEPs were obtained from the transcriptomic and proteomic study respectively. 14 common genes were identified, which is enriched in muscle contraction and inflammation-related pathways. Meanwhile, we observed 33 significant differences in the infiltration of cells in DMD. Afterwards, a total of 22 miRNAs and 23 TF genes interacted with the common genes, including TFAP2C, MAX, MYC, NFKB1, RELA, hsa-miR-1255a, hsa-miR-130a, hsa-miR-130b, hsa-miR-152, and hsa-miR-17. In addition, three genes (ATP6AP2, CTSS, and VIM) showed excellent diagnostic performance on discriminating DMD in GSE1004, GSE3307, GSE6011 and GSE38417 datasets (all AUC > 0.8), which is validated in patients (10 DMD vs. 10 controls), DMD with exon 55 mutations, mdx mouse, and nomogram model. Conclusion Taken together, ATP6AP2, CTSS, and VIM play important roles in the inflammatory response in DMD, which may serve as diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xuan Wu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China,Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Nan Dong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Liqiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meirong Liu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianhua Jiang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tieyu Tang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Hongru Zhao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China,*Correspondence: Hongru Zhao, ; Qi Fang,
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China,*Correspondence: Hongru Zhao, ; Qi Fang,
| |
Collapse
|
13
|
Liu X, Zhao W, Shu S, Zhang W. Duchenne muscular dystrophy involves the myocardium and causes arrhythmia: Case report. Front Cardiovasc Med 2022; 9:974843. [PMID: 36440017 PMCID: PMC9681897 DOI: 10.3389/fcvm.2022.974843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022] Open
Abstract
Background Patients with muscular dystrophy have mutations in the gene that can lead to severe muscle wasting, respiratory issues or heart failure between ages 30 and 40. Currently, there is no effective treatment for DMD-induced heart failure. Case presentation We report a patient with recurrent unexplained fever and muscle soreness was definitely diagnosed with DMD. An analysis of the patient's genetics revealed a nonsense mutation (C.1207G > T). His DMD was treated with hormones. Also, the patient's fever is under control because of hormone therapy. However, as the disease progresses, the heart structure and function gradually change, and eventually malignant arrhythmias occur. Conclusion We report a rare case of DMD involving the heart causing heart failure and malignant arrhythmia. Currently, no complete treatment is available for these patients, but our treatment regimen may benefit our patient and improve his outcomes.
Collapse
|
14
|
Szabo SM, Klimchak AC, Qian C, Iannaccone S, Popoff E, Gooch KL. Characterizing the Occurrence of Key Clinical Milestones in Duchenne Muscular Dystrophy in the United States Using Real-World Data. J Neuromuscul Dis 2022; 9:689-699. [DOI: 10.3233/jnd-220816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Background: Data on the clinical course of Duchenne muscular dystrophy (DMD) exist from well-characterized clinical cohorts but estimates from real-world populations are fewer. Objective: The objective was to estimate the prevalence of key clinical milestones by age, among real-world commercially-insured DMD patients in the United States. Methods: MarketScan claims (2013–2018) were used to identify males with DMD. The percentages with wheelchair use or experiencing scoliosis, neurologic/neuropsychiatric involvement, cardiomyopathy, and respiratory involvement were tabulated; as were the median (interquartile range [IQR]) ages at first observed occurrence within the claims data. Results: Among DMD patients (n = 1,964), the median (IQR) baseline age was 15 (9–21) years, and median follow-up was 1.7 years. Wheelchair use was observed in 55% of those aged 8 to 13 years at cohort entry; scoliosis, among 38% of those 8 to 10 and 52% of those 11 to 13 years; neurologic/neuropsychiatric involvement, among 41–43% of those 8 to 13 years; respiratory involvement, among 45% of those 14 to 19 years; and cardiomyopathy, among 68% of those 14 to 16 and 58% of those 17 to 19 years. Conclusions: The prevalence of key clinical milestones across ages was broadly consistent with published findings. Variability in estimates reflect clinical heterogeneity; these contemporary estimates from real-world data help characterize clinical outcomes in DMD.
Collapse
Affiliation(s)
- Shelagh M. Szabo
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | | | - Christina Qian
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | - Susan Iannaccone
- University of Texas Southwestern, Harry Hines Blvd, Dallas TX, USA
| | - Evan Popoff
- Broadstreet Health Economics & Outcomes Research, Vancouver BC, Canada
| | | |
Collapse
|
15
|
Szűcs Z, Pinti É, Haltrich I, Szén OP, Nagy T, Barta E, Méhes G, Bidiga L, Török O, Ujfalusi A, Koczok K, Balogh I. An Ultra-Rare Manifestation of an X-Linked Recessive Disorder: Duchenne Muscular Dystrophy in a Female Patient. Int J Mol Sci 2022; 23:ijms232113076. [PMID: 36361862 PMCID: PMC9655586 DOI: 10.3390/ijms232113076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 11/30/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common inherited muscle dystrophy. Patients are characterized by muscle weakness, gross motor delay, and elevated serum creatinine kinase (CK) levels. The disease is caused by mutations in the DMD gene located on the X chromosome. Due to the X-linked recessive inheritance pattern, DMD most commonly affects males, who are generally diagnosed between the age of 3–5 years. Here we present an ultra-rare manifestation of DMD in a female patient. Cytogenetic examination showed that she has a t(X;10)(p21.1;p12.1) translocation, which turned out to affect the DMD gene with one of the breakpoints located in exon 54 (detected by genome sequencing). The X-inactivation test revealed skewed X-inactivation (ratio 99:1). Muscle histology and dystrophin immunohistochemistry showed severe dystrophic changes and highly reduced dystrophin expression, respectively. These results, in accordance with the clinical picture and a highly elevated serum CK, led to the diagnosis of DMD. In conclusion, although in very rare cases, DMD can manifest in female patients as well. In this case, a balanced X-autosome reciprocal translocation disrupts the DMD gene and skewed X-inactivation leads to the manifestation of the DMD phenotype.
Collapse
Affiliation(s)
- Zsuzsanna Szűcs
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Éva Pinti
- 2nd Department of Pediatrics, Semmelweis University, 1094 Budapest, Hungary
| | - Irén Haltrich
- 2nd Department of Pediatrics, Semmelweis University, 1094 Budapest, Hungary
| | - Orsolya Pálné Szén
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tibor Nagy
- Bioinformatics and Functional Genome Analysis Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Endre Barta
- Bioinformatics and Functional Genome Analysis Research Group, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Department of Genetics and Genomics, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - László Bidiga
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Olga Török
- Medical and Health Science Centre, Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Anikó Ujfalusi
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Katalin Koczok
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (K.K.); (I.B.)
| | - István Balogh
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Correspondence: (K.K.); (I.B.)
| |
Collapse
|
16
|
Wein N, Vetter TA, Vulin A, Simmons TR, Frair EC, Bradley AJ, Gushchina LV, Almeida CF, Huang N, Lesman D, Rajakumar D, Weiss RB, Flanigan KM. Systemic delivery of an AAV9 exon-skipping vector significantly improves or prevents features of Duchenne muscular dystrophy in the Dup2 mouse. Mol Ther Methods Clin Dev 2022; 26:279-293. [PMID: 35949298 PMCID: PMC9356240 DOI: 10.1016/j.omtm.2022.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022]
Abstract
Duchenne muscular dystrophy (DMD) is typically caused by mutations that disrupt the DMD reading frame, but nonsense mutations in the 5′ part of the gene induce utilization of an internal ribosomal entry site (IRES) in exon 5, driving expression of a highly functional N-truncated dystrophin. We have developed an AAV9 vector expressing U7 small nuclear RNAs targeting DMD exon 2 and have tested it in a mouse containing a duplication of exon 2, in which skipping of both exon 2 copies induces IRES-driven expression, and skipping of one copy leads to wild-type dystrophin expression. One-time intravascular injection either at postnatal days 0–1 or at 2 months results in efficient exon skipping and dystrophin expression, and significant protection from functional and pathologic deficits. Immunofluorescence quantification showed 33%–53% average dystrophin intensity and 55%–79% average dystrophin-positive fibers in mice treated in adulthood, with partial amelioration of DMD pathology and correction of DMD-associated alterations in gene expression. In mice treated neonatally, dystrophin immunofluorescence reached 49%–85% of normal intensity and 76%–99% dystrophin-positive fibers, with near-complete correction of dystrophic pathology, and these beneficial effects persisted for at least 6 months. Our results demonstrate the robustness, durability, and safety of exon 2 skipping using scAAV9.U7snRNA.ACCA, supporting its clinical use.
Collapse
Affiliation(s)
- Nicolas Wein
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Tatyana A Vetter
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Adeline Vulin
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Tabatha R Simmons
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Emma C Frair
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Adrienne J Bradley
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Liubov V Gushchina
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Camila F Almeida
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Nianyuan Huang
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Daniel Lesman
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Dhanarajan Rajakumar
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Robert B Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Abigail Wexner Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA.,Department of Neurology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Guo D, Li X, Liu N, Yu X, Shu J, Sheng W, Li D, Cai C. Beware of missed diagnosis in patients with multiple genetic diseases: a case report. BMC Pediatr 2022; 22:436. [PMID: 35858850 PMCID: PMC9297618 DOI: 10.1186/s12887-022-03490-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/12/2022] [Indexed: 11/10/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked recessive inherited disorder caused by the absence of the Dystrophin protein. Cerebral cavernous malformations (CCMs) are the most common vascular abnormalities in the central nervous system caused by the absence of the products of the CCM genes. Most CCMs cases reported occurring in a sporadic form are often asymptomatic. Case presentation We report a rare case of a 7-year-old Chinese boy with a co-existing DMD and sporadic CCMs. We found classic clinical features of DMD and non-specific pathological changes in his brain. We made the definitive diagnosis based on the results of whole-exome sequencing (WES), a repeat from exon 3 to exon 9 of the DMD inherited from his mother, and a de novo heterozygote nonsense mutation C.418G > T of the PDCD10 exon 6. Conclusion We should take care to avoid missed diagnoses in patients with multiple genetic disorders.
Collapse
Affiliation(s)
- Detong Guo
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Graduate College of Tianjin Medical University, Tianjin, 300070, China
| | - Xuemei Li
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Department of Neurology, Tianjin Children's Hospital, Beichen District, No. 238 Longyan Road, Tianjin, 300134, China
| | - Nan Liu
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Tianjin Pediatric Research Institute, Tianjin, 300134, China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China
| | - Xiaoli Yu
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Department of Neurology, Tianjin Children's Hospital, Beichen District, No. 238 Longyan Road, Tianjin, 300134, China
| | - Jianbo Shu
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Tianjin Pediatric Research Institute, Tianjin, 300134, China.,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China
| | - Wenchao Sheng
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.,Graduate College of Tianjin Medical University, Tianjin, 300070, China
| | - Dong Li
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China. .,Department of Neurology, Tianjin Children's Hospital, Beichen District, No. 238 Longyan Road, Tianjin, 300134, China.
| | - Chunquan Cai
- Tianjin Children's Hospital (Children's Hospital of Tianjin University), Beichen District, No. 238 Longyan Road, Tianjin, 300134, China. .,Tianjin Pediatric Research Institute, Tianjin, 300134, China. .,Tianjin Key Laboratory of Birth Defects for Prevention and Treatment, Tianjin, 300134, China. .,Department of Neurosurgery, Tianjin Children's Hospital, Tianjin, 300134, China.
| |
Collapse
|
18
|
Szabo SM, Audhya IF, Rogula B, Feeny D, Gooch KL. Factors associated with the health-related quality of life among people with Duchenne muscular dystrophy: a study using the Health Utilities Index (HUI). Health Qual Life Outcomes 2022; 20:93. [PMID: 35690783 PMCID: PMC9188127 DOI: 10.1186/s12955-022-02001-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Background Data on health state utility in Duchenne muscular dystrophy (DMD) are few. This study estimated mean utility values by age, ambulatory status and over time, and investigated which aspects of health-related quality-of-life (HRQoL) are most strongly associated with utility in DMD. Methods Data from placebo-treated ambulant boys with DMD with exon 51 skip amenable mutations, (NCT01254019), were included. Ambulatory function assessments were conducted at baseline and every 12 weeks for the trial duration. Family member proxies completed the Health Utility Index (HUI) at baseline, 24 and 48 weeks; and HUI3 and HUI2 utility values were summarized. Changes in HUI attribute level over time, and predictors of changes in utility, were explored. Results Sixty-one boys (mean [range] age of 8.0 [5–16] years) were included in the analysis. Mean baseline utilities were 0.82 (HUI3) and 0.87 (HUI2); and utilities were 0.35 (HUI3) and 0.55 (HUI2) after loss of ambulation (LOA, where applicable). Over the follow-up period mean utility declined more among the older versus younger boys. Pain accounted for the highest proportion of variability (42%) in change in HUI3 utility from baseline to week 48, while for HUI2, self-care (39%) did. After LOA, change in ambulation levels explained 88% of the decline in mean HUI3 utility and change in mobility levels explained 66% of the decline in mean HUI2 utility. Conclusions Utility values among this sample were higher than previously published estimates. In younger boys utility remained relatively stable, but older boys and those losing ambulation experienced important declines over follow-up.
Supplementary Information The online version contains supplementary material available at 10.1186/s12955-022-02001-0.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, Canada.
| | - Ivana F Audhya
- Sarepta Therapeutics, 215 First St, Cambridge, MA, 02142, USA
| | - Basia Rogula
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, Canada
| | - David Feeny
- McMaster University and Health Utilities Inc., Hamilton, ON, Canada
| | | |
Collapse
|
19
|
Laurila PP, Luan P, Wohlwend M, Zanou N, Crisol B, Imamura de Lima T, Goeminne LJE, Gallart-Ayala H, Shong M, Ivanisevic J, Place N, Auwerx J. Inhibition of sphingolipid de novo synthesis counteracts muscular dystrophy. SCIENCE ADVANCES 2022; 8:eabh4423. [PMID: 35089797 PMCID: PMC8797791 DOI: 10.1126/sciadv.abh4423] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 11/01/2021] [Indexed: 05/29/2023]
Abstract
Duchenne muscular dystrophy (DMD), the most common muscular dystrophy, is a severe muscle disorder, causing muscle weakness, loss of independence, and premature death. Here, we establish the link between sphingolipids and muscular dystrophy. Transcripts of sphingolipid de novo biosynthesis pathway are up-regulated in skeletal muscle of patients with DMD and other muscular dystrophies, which is accompanied by accumulation of metabolites of the sphingolipid pathway in muscle and plasma. Pharmacological inhibition of sphingolipid synthesis by myriocin in the mdx mouse model of DMD ameliorated the loss in muscle function while reducing inflammation, improving Ca2+ homeostasis, preventing fibrosis of the skeletal muscle, heart, and diaphragm, and restoring the balance between M1 and M2 macrophages. Myriocin alleviated the DMD phenotype more than glucocorticoids. Our study identifies inhibition of sphingolipid synthesis, targeting multiple pathogenetic pathways simultaneously, as a strong candidate for treatment of muscular dystrophies.
Collapse
Affiliation(s)
- Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Peiling Luan
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Barbara Crisol
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ludger J. E. Goeminne
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Minho Shong
- Research Center for Endocrine and Metabolic Diseases, Chungnam National University Hospital, Chungnam National University School of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Department of Physiology, Faculty of Biology-Medicine, University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
20
|
MUNNO PAULAM, BARROSO POLIANAR, VASCONCELOS BRUNAF, SILVA GEOVANNEBDA, SALGUEIRO THIAGOM, AGUIAR HELOISAH, VITORINO LETÍCIAM, OLIVEIRA MURILOX, MARTINS HELENR, GAIAD THAISP, MACHADO ALEXS. Acute toxicity and regenerative dose finding of an extract of Miconia ferruginata DC. in a mouse model of Duchenne muscular dystrophy. AN ACAD BRAS CIENC 2022; 94:e20210190. [DOI: 10.1590/0001-3765202220210190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/02/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- PAULA M. MUNNO
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | | | | | | | | | | | | | | | - HELEN R. MARTINS
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | - THAIS P. GAIAD
- Universidade Federal dos Vales do Jequitinhonha e Mucuri, Brazil
| | | |
Collapse
|
21
|
Szabo SM, Gooch KL, Mickle AT, Salhany RM, Connolly AM. The impact of genotype on outcomes in individuals with Duchenne muscular dystrophy: A systematic review. Muscle Nerve 2021; 65:266-277. [PMID: 34878187 DOI: 10.1002/mus.27463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/07/2022]
Abstract
Duchenne muscular dystrophy (DMD) is associated with progressive muscle weakness, loss of ambulation (LOA), and early mortality. In this review we have synthesized published data on the clinical course of DMD by genotype. Using a systematic search implemented in Medline and Embase, 53 articles were identified that describe the clinical course of DMD, with pathogenic variants categorizable by exon skip or stop-codon readthrough amenability and outcomes presented by age. Outcomes described included those related to ambulatory, cardiac, pulmonary, or cognitive function. Estimates of the mean (95% confidence interval) age at LOA ranged from 9.1 (8.7-9.6) years among 90 patients amenable to skipping exon 53 to 11.5 (9.5-13.5) years among three patients amenable to skipping exon 8. Although function worsened with age, the impact of genotype was less clear for other outcomes (eg, forced vital capacity and left ventricular ejection fraction). Understanding the distribution of pathogenic variants is important for studies in DMD, as this research suggests major differences in the natural history of disease. In addition, specific details of the use of key medications, including corticosteroids, antisense oligonucleotides, and cardiac medications, should be reported.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Alexis T Mickle
- Broadstreet Heath Economics & Outcomes Research, Vancouver, British Columbia, Canada
| | | | - Anne M Connolly
- Division of Neurology, Nationwide Children's Hospital, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
22
|
Lazovic M, Nikolic D, Boyer FC, Borg K, Ceravolo MG, Zampolini M, Kiekens C. Evidence based position paper on Physical and Rehabilitation Medicine practice for people with muscular dystrophies. Eur J Phys Rehabil Med 2021; 57:1036-1044. [PMID: 34823337 DOI: 10.23736/s1973-9087.21.07121-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Muscular dystrophies present a group of inherited degenerative disorder that are characterized by progressive muscular weakness. This evidence-based position paper represents the official position of the European Union through the UEMS PRM Section. The aim of the paper is to evaluate the role of the physical and rehabilitation medicine (PRM) physician and PRM practice for people with muscular dystrophies. A systematic review of the literature and a consensus procedure by means of a Delphi process have been performed involving the delegates of all European countries represented in the UEMS PRM Section. The systematic literature review is reported together with thirty-three recommendations resulting from the Delphi procedure. The role of the PRM physician is to assess the functional status of persons with muscular dystrophy and to plan, monitor and lead PRM program in an interdisciplinary setting within a multiprofessional team.
Collapse
Affiliation(s)
- Milica Lazovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia - .,Institute for Rehabilitation, Belgrade, Serbia -
| | - Dejan Nikolic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Department of Physical Medicine and Rehabilitation, University Children's Hospital, Belgrade, Serbia
| | - François C Boyer
- PRM Department, Sebastopol Hospital, Reims Champagne Ardenne University, Reims, France
| | - Kristian Borg
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Maria G Ceravolo
- Department of Experimental and Clinical Medicine, Neurorehabilitation Clinic, University Hospital of Ancona, Politecnica delle Marche University, Ancona, Italy
| | - Mauro Zampolini
- Department of Rehabilitation, San Giovanni Battista Hospital, Foligno, Perugia, Italy
| | | |
Collapse
|
23
|
De Palma FDE, Nunziato M, D’Argenio V, Savarese M, Esposito G, Salvatore F. Comprehensive Molecular Analysis of DMD Gene Increases the Diagnostic Value of Dystrophinopathies: A Pilot Study in a Southern Italy Cohort of Patients. Diagnostics (Basel) 2021; 11:diagnostics11101910. [PMID: 34679607 PMCID: PMC8534830 DOI: 10.3390/diagnostics11101910] [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: 08/20/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Duchenne/Becker muscular dystrophy (DMD/BMD) is an X-linked neuromuscular disease due to pathogenic sequence variations in the dystrophin (DMD) gene, one of the largest human genes. More than 70% of DMD gene defects result from genomic rearrangements principally leading to large deletions, while the remaining are small nucleotide variants, including nonsense and missense variants, small insertions/deletions or splicing alterations. Considering the large size of the gene and the wide mutational spectrum, the comprehensive molecular diagnosis of DMD/BMD is complex and may require several laboratory methods, thus increasing the time and costs of the analysis. In an attempt to simplify DMD/BMD molecular diagnosis workflow, we tested an NGS method suitable for the detection of all the different types of genomic variations that may affect the DMD gene. Forty previously analyzed patients were enrolled in this study and re-analyzed using the next generation sequencing (NGS)-based single-step procedure. The NGS results were compared with those from multiplex ligation-dependent probe amplification (MLPA)/multiplex PCR and/or Sanger sequencing. Most of the previously identified deleted/duplicated exons and point mutations were confirmed by NGS and 1 more pathogenic point mutation (a nonsense variant) was identified. Our results show that this NGS-based strategy overcomes limitations of traditionally used methods and is easily transferable to routine diagnostic procedures, thereby increasing the diagnostic power of DMD molecular analysis.
Collapse
Affiliation(s)
- Fatima Domenica Elisa De Palma
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Marcella Nunziato
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Valeria D’Argenio
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Via di Val Cannuta 247, 00166 Roma, Italy
| | - Maria Savarese
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
| | - Gabriella Esposito
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: (G.E.); (F.S.); Tel.: +81-746-3146 (G.E.); +81-373-7826 (F.S.)
| | - Francesco Salvatore
- CEINGE-Biotecnologie Avanzate s.c.ar.l., 80145 Naples, Italy; (F.D.E.D.P.); (M.N.); (V.D.); (M.S.)
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80131 Naples, Italy
- Correspondence: (G.E.); (F.S.); Tel.: +81-746-3146 (G.E.); +81-373-7826 (F.S.)
| |
Collapse
|
24
|
Klimchak AC, Szabo SM, Qian C, Popoff E, Iannaccone S, Gooch KL. Characterizing demographics, comorbidities, and costs of care among populations with Duchenne muscular dystrophy with Medicaid and commercial coverage. J Manag Care Spec Pharm 2021; 27:1426-1437. [PMID: 34595954 PMCID: PMC10391028 DOI: 10.18553/jmcp.2021.27.10.1426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND: Duchenne muscular dystrophy (DMD) is a severe X-linked progressive neurodegenerative disease characterized by loss of ambulation, cardiomyopathy, respiratory insufficiency, and early mortality. Few data are available that describe the direct medical costs among patients with DMD in the United States. OBJECTIVE: To characterize the demographics, comorbidity burden, and direct monthly costs of care among patients with DMD with Medicaid and with commercial insurance coverage. METHODS: IBM MarketScan Commercial and Multi-State Medicaid claims (2013-2018) were used to identify males aged 30 years or under with diagnostic codes for muscular dystrophy or DMD; additional exclusion criteria were applied to identify those with probable DMD. Baseline characteristics and comorbidities were tabulated. The frequency of health care resource use and median (interquartile range [IQR]) monthly costs (in 2018 USD) were estimated from those with at least 12 months of continuous follow-up. RESULTS: Median (IQR) baseline ages were similar between the Medicaid (14 [9-20] years; n = 2,007) and commercial (15 [9-21] years; n = 1,964) DMD cohorts. The frequency of comorbidities over the period was slightly higher with those on Medicaid. The median duration of follow-up was 3.1 years among members of the Medicaid DMD cohort and 1.7 years among the commercial DMD cohort. Median monthly resource use was generally higher among the Medicaid DMD cohort; nonetheless, median (IQR) monthly costs were similar at $1,735 ($367-$5,281) for the Medicaid DMD cohort vs $1,883 ($657-$6,796) for the commercial DMD cohort. CONCLUSIONS: The demographic characteristics and median direct medical costs were similar between patients with commercial vs Medicaid coverage, even though patients with Medicaid coverage had higher resource use. Despite challenges in definitively identifying DMD patients using claims data, these findings help characterize contemporary DMD populations in the United States and the related direct economic burden to the payer. DISCLOSURES: This study was funded by Sarepta Therapeutics, Inc. Klimchak and Gooch are employees of Sarepta Therapeutics Inc. Szabo, Qian, and Popoff are employees of Broadstreet HEOR, which received funds from Sarepta Therapeutics, Inc., for work on this study. Iannaccone has received research funding or consulting fees from Avexis, Biogen, Fibrogen, Mallinkrodt, Regeneron, Sarepta Therapeutics, Inc., Scholar Rock, PTC Therapeutics, Pfizer, MDA, CureSMA, NIH, Genentech-Roche, and BCBS. Publication of the study results was not contingent on the sponsor's approval or censorship of the manuscript. Information from this study was presented, in part, at the AMCP Virtual Annual Meeting, April 21-24, 2020.
Collapse
Affiliation(s)
| | | | | | | | - Susan Iannaccone
- Children's Medical Center Ambulatory Care Pavilion, University of Texas Southwestern Medical Center, Dallas
| | | |
Collapse
|
25
|
Szabo SM, Salhany RM, Deighton A, Harwood M, Mah J, Gooch KL. The clinical course of Duchenne muscular dystrophy in the corticosteroid treatment era: a systematic literature review. Orphanet J Rare Dis 2021; 16:237. [PMID: 34022943 PMCID: PMC8141220 DOI: 10.1186/s13023-021-01862-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/10/2021] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is a severe rare progressive inherited neuromuscular disorder, leading to loss of ambulation (LOA) and premature mortality. The standard of care for patients with DMD has been treatment with corticosteroids for the past decade; however a synthesis of contemporary data describing the clinical course of DMD is lacking. The objective was to summarize age at key clinical milestones (loss of ambulation, scoliosis, ventilation, cardiomyopathy, and mortality) in the corticosteroid-treatment-era. Methods A systematic review was conducted using MEDLINE and EMBASE. The percentage experiencing key clinical milestones, and the mean or median age at those milestones, was synthesized from studies from North American populations, published between 2007 and 2018. Results From 5637 abstracts, 29 studies were included. Estimates of the percentage experiencing key clinical milestones, and age at those milestones, showed heterogeneity. Up to 30% of patients lost ambulation by age 10 years, and up to 90% by 15 years of age. The mean age at scoliosis onset was approximately 14 years. Ventilatory support began from 15 to 18 years, and up to half of patients required ventilation by 20 years of age. Registry-based estimates suggest that 70% had evidence of cardiomyopathy by 15 years and almost all by 20 years of age. Finally, mortality rates up to 16% by age 20 years were reported; among those surviving to adulthood mortality was up to 60% by age 30 years. Conclusions Contemporary natural history studies from North America report that LOA on average occurs in the early teens, need for ventilation and cardiomyopathy in the late teens, and death in the third or fourth decade of life. Variability in rates may be due to differences in study design, treatment with corticosteroids or other disease-modifying agents, variations in clinical practices, and dystrophin mutations. Despite challenges in synthesizing estimates, these findings help characterize disease progression among contemporary North American DMD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-021-01862-w.
Collapse
Affiliation(s)
- Shelagh M Szabo
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada.
| | - Renna M Salhany
- Sarepta Therapeutics, 215 First St, Cambridge, MA, 02142, USA
| | - Alison Deighton
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - Meagan Harwood
- Broadstreet HEOR, 201 - 343 Railway St, Vancouver, BC, V6A 1A4, Canada
| | - Jean Mah
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
26
|
Xu X, Hao Y, Wu J, Zhao J, Xiong S. Assessment of Weighted Gene Co-Expression Network Analysis to Explore Key Pathways and Novel Biomarkers in Muscular Dystrophy. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:431-444. [PMID: 33883925 PMCID: PMC8053709 DOI: 10.2147/pgpm.s301098] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/23/2021] [Indexed: 12/12/2022]
Abstract
Purpose This study aimed to explore the key molecular pathways involved in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) and thereby identify hub genes to be potentially used as novel biomarkers using a bioinformatics approach. Methods Raw GSE109178 data were collected from the Gene Expression Omnibus (GEO) database. Weighted gene co-expression network analysis (WGCNA) was conducted on the top 50% of altered genes. The key modules associated with the clinical features of DMD and BMD were identified. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed using the DAVID website. A protein-protein interaction (PPI) network was constructed using the STRING website. MCODE, together with the Cytohubba plug-ins of Cytoscape, screened out the potential hub genes, which were subsequently verified via receiver operating characteristic (ROC) curves in other datasets. Results Among the 11 modules obtained, the black module was predominantly associated with pathology and DMD, whereas the light-green module was primarily related to age and BMD. Functional enrichment assessments indicated that the genes in the black module were primarily clustered in “immune response” and “phagosome,” whereas the ones in the light-green module were chiefly enriched in “protein polyubiquitination”. Eleven essential genes were eventually identified, including VCAM1, TYROBP, CD44, ITGB2, CSF1R, LCP2, C3AR1, CCL2, and ITGAM for DMD, along with UBA5 and UBR2 for BMD. Conclusion Overall, our findings may be useful for investigating the mechanisms underlying DMD and BMD. In addition, the hub genes discovered might serve as novel molecular markers correlated with dystrophinopathies.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yuehan Hao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Jiao Wu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, People's Republic of China
| | - Jing Zhao
- Department of Neurology, The First Hospital of China Medical University, Shenyang, People's Republic of China
| | - Shuang Xiong
- Liaoning Academy of Analytic Science, Construction Engineering Center of Important Technology Innovation and Research and Development Base in Liaoning Province, Shenyang, People's Republic of China
| |
Collapse
|
27
|
Abstract
Increased understanding of disease pathophysiology and advances in gene therapies and drug technologies are revolutionizing treatment of muscular dystrophies and motor neuron disorders (MNDs). New drugs have been approved for Duchenne muscular dystrophy, spinal muscular atrophy, and amyotrophic lateral sclerosis. For other diseases, new targets have been identified, and new therapies are in clinical trials. The impact of such therapies will be fully understood only in the next decades. Cost burden and accessibility are major challenges in the wide application of new drugs. This article reviews advances in gene therapies, newly approved drugs, and therapeutic promises in muscular dystrophies and MNDs.
Collapse
|
28
|
Wang Y, Chen Y, Wang SM, Liu X, Gu YN, Feng Z. Prenatal diagnosis of Duchenne muscular dystrophy revealed a novel mosaic mutation in Dystrophin gene: a case report. BMC MEDICAL GENETICS 2020; 21:222. [PMID: 33176713 PMCID: PMC7661169 DOI: 10.1186/s12881-020-01157-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/25/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND Duchenne muscular dystrophies (DMDs) are X-linked recessive neuromuscular disorders with malfunction or absence of the Dystrophin protein. Precise genetic diagnosis is critical for proper planning of patient care and treatment. In this study, we described a Chinese family with mosaic DMD mutations and discussed the best method for prenatal diagnosis and genetic counseling of X-linked familial disorders. METHODS We investigated all variants of the whole dystrophin gene using multiple DNA samples isolated from the affected family and identified two variants of the DMD gene in a sick boy and two female carriers by targeted next generation sequencing (TNGS), Sanger sequencing, and haplotype analysis. RESULTS We identified the hemizygous mutation c.6794delG (p.G2265Efs*6) of DMD in the sick boy, which was inherited from his mother. Unexpectedly, a novel heterozygous mutation c.6796delA (p.I2266Ffs*5) of the same gene, which was considered to be a de novo variant, was detected from his younger sister instead of his mother by Sanger sequencing. However, further NGS analysis of the mother and her amniotic fluid samples revealed that the mother carried a low-level mosaic c.6796delA mutation. CONCLUSIONS We reported two different mutations of the DMD gene in two siblings, including the novel mutation c.6796delA (p.I2266Ffs*5) inherited from the asymptomatic mosaic-carrier mother. This finding has enriched the knowledge of the pathogenesis of DMD. If no mutation is detected in obligate carriers, the administration of intricate STR/NGS/Sanger analysis will provide new ideas on the prenatal diagnosis of DMD.
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China. .,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China. .,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China. .,Clinical Biobank Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yuhan Chen
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China.,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China.,Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100700, China
| | - San Mei Wang
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China.,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China.,Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100700, China
| | - Xin Liu
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China.,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China.,Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100700, China
| | - Ya Nan Gu
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China.,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China.,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China.,Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100700, China
| | - Zhichun Feng
- Department of Clinical Genetics, BaYi Children's Hospital, Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, China. .,National Engineering Laboratory for Birth defects prevention and control of key technology, Beijing, 100700, China. .,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, 100700, China. .,Department of Pediatrics, Chinese PLA General Hospital, Beijing, 100700, China.
| |
Collapse
|
29
|
Iftikhar M, Frey J, Shohan MJ, Malek S, Mousa SA. Current and emerging therapies for Duchenne muscular dystrophy and spinal muscular atrophy. Pharmacol Ther 2020; 220:107719. [PMID: 33130193 DOI: 10.1016/j.pharmthera.2020.107719] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
Many neuromuscular diseases are genetically inherited or caused by mutations in motor function proteins. Two of the most prevalent neuromuscular diseases are Duchenne Muscular Dystrophy (DMD) and Spinal Muscular Atrophy (SMA), which are often diagnosed during the early years of life, contributing to life-long debilitation and shorter longevity. DMD is caused by mutations in the dystrophin gene resulting in critical muscle wasting, with cardiac or respiratory failure by age 30. Lack of dystrophin protein is the leading cause of degeneration of skeletal and cardiac muscle. Corticosteroids and artificial respirators remain as the gold-standard management of complications and have significantly extended the life span of these patients. Additionally, drug therapies including eteplirsen (EXONDYS 51®), golodirsen (VYONDYS 53™), and viltolarsen (VILTEPSO®) have been approved by the FDA to treat specific types of DMD. SMA is defined by the degeneration of the anterior horn cells in the spinal cord and destruction of motor neuron nuclei in the lower brain-stem caused by SMN1 gene deletion. Loss of SMN1 protein is partly compensated by SMN2 protein synthesis with disease severity being affected by the success of SMN2 gene synthesis. Evidence-based recommendations for SMA are directed towards supportive therapy and providing adequate nutrition and respiratory assistance as needed. Treatment and prevention of complications of muscle weakness are crucial for reducing the phenotype expression of SMA. Furthermore, drug therapies including injectables such as onasemnogene abeparvovec-xioi (ZOLGENSMA®), nusinersen (SPINRAZA®), and an oral-solution, risdiplam (EVRYSDI™), are medications that have been FDA-approved for the treatment of SMA. This review discusses the current and emerging therapeutic options for patients with DMD and SMA.
Collapse
Affiliation(s)
- Mohsan Iftikhar
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States of America
| | - Justin Frey
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States of America
| | - Md Jasimuddin Shohan
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States of America
| | - Sohail Malek
- Department of Pediatric Neurology, Albany Medical Center, Albany, NY 12208, United States of America
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States of America.
| |
Collapse
|
30
|
Genotype and age at diagnosis in Thai boys with Duchenne muscular dystrophy (DMD). Neuromuscul Disord 2020; 30:839-844. [PMID: 32962870 DOI: 10.1016/j.nmd.2020.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/04/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022]
Abstract
Gene-based therapy is a treatment for Duchenne muscular dystrophy (DMD) has become lately available; limited use for specific of mutation and percentages of the patients. Diagnosis in Thailand is made by muscle biopsy or multiplex ligation-dependent probe amplification (MLPA). Appropriate treatment in developing countries is difficult because gene sequencing is expensive and has limited availability. We aimed to identify the clinical and genetic characteristics of Thai DMD. Patients aged 0-22 years were recruited from the pediatric neuromuscular clinic of Siriraj Hospital during 2017-2019. Ninety-four charts were reviewed for clinical and laboratory data. Patients with negative MLPA who underwent next generation sequencing were consented. The mean age at onset and diagnosis was 4 and 7 years, respectively. Approximately 70% of patients had loss of ambulation by the mean age of 9.6 ± 1.8 years. Eighty percent were treated with glucocorticoids. Genetic testing was performed in 70 patients. Molecular analysis revealed mutations in 90% of cases, including exon deletions in 48.57%, nonsense mutations in 20%, frameshift mutations in 12.86%, splice site in 7.14%, exon duplications in 5.71%, and in-frame deletion in 2.86%. Gene sequencing should be performed because baseline genetic mutation data is essential for gene-based therapies that will become available in the future.
Collapse
|
31
|
El Kadiri Y, Selouani Y, Ratbi I, Lyahyai J, Zrhidri A, Sahli M, Ouhenach M, Jaouad IC, Sefiani A, Sbiti A. Molecular diagnosis of dystrophinopathies in Morocco and report of six novel mutations. Clin Chim Acta 2020; 506:28-32. [PMID: 32169422 DOI: 10.1016/j.cca.2020.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 10/24/2022]
Abstract
Dystrophinopathies are the most common genetic neuromuscular disorders during childhood, with an X-linked recessive inheritance pattern. Because of clinical and genetic heterogeneity of dystrophinopathies, genetic testing of dystrophin gene at Xp21.2 is constantly evolving. Multiplex Polymerase Chain Reaction (MPCR) is used in the first line to detect common exon deletions of dystrophin gene (accounting for 65% of mutations), followed by the Multiplex Ligation-dependent Probe Amplification (MLPA) technique to reveal deletions of exons outside the usual hotspot and duplications in male and female carriers. (MLPA adds another 10-15% positive cases to MPCR). Recently, Next Generation Sequencing allows to screen for rare large and point mutations. We report here, molecular analysis results of dystrophin gene during 27 years in a large Moroccan cohort of 356 patients, using the multiplex polymerase chain reaction (MPCR) to screen for hot-spot exon deletions. First applications of whole dystrophin gene sequencing in our lab lead to the identification of six novel mutations.
Collapse
Affiliation(s)
- Youssef El Kadiri
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco.
| | - Yassir Selouani
- Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Ilham Ratbi
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Jaber Lyahyai
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco
| | - Abdelali Zrhidri
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Maryem Sahli
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Mouna Ouhenach
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Imane Cherkaoui Jaouad
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Abdelaziz Sefiani
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Morocco; Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| | - Aziza Sbiti
- Department of Medical Genetics, National Institute of Health in Rabat, BP 769 Agdal, 10 090 Rabat, Morocco
| |
Collapse
|
32
|
How the central domain of dystrophin acts to bridge F-actin to sarcolemmal lipids. J Struct Biol 2019; 209:107411. [PMID: 31689503 DOI: 10.1016/j.jsb.2019.107411] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/07/2019] [Accepted: 10/29/2019] [Indexed: 01/08/2023]
Abstract
Dystrophin is a large intracellular protein that prevents sarcolemmal ruptures by providing a mechanical link between the intracellular actin cytoskeleton and the transmembrane dystroglycan complex. Dystrophin deficiency leads to the severe muscle wasting disease Duchenne Muscular Dystrophy and the milder allelic variant, Becker Muscular Dystrophy (DMD and BMD). Previous work has shown that concomitant interaction of the actin binding domain 2 (ABD2) comprising spectrin like repeats 11 to 15 (R11-15) of the central domain of dystrophin, with both actin and membrane lipids, can greatly increase membrane stiffness. Based on a combination of SAXS and SANS measurements, mass spectrometry analysis of cross-linked complexes and interactive low-resolution simulations, we explored in vitro the molecular properties of dystrophin that allow the formation of ABD2-F-actin and ABD2-membrane model complexes. In dystrophin we identified two subdomains interacting with F-actin, one located in R11 and a neighbouring region in R12 and another one in R15, while a single lipid binding domain was identified at the C-terminal end of R12. Relative orientations of the dystrophin central domain with F-actin and a membrane model were obtained from docking simulation under experimental constraints. SAXS-based models were then built for an extended central subdomain from R4 to R19, including ABD2. Overall results are compatible with a potential F-actin/dystrophin/membrane lipids ternary complex. Our description of this selected part of the dystrophin associated complex bridging muscle cell membrane and cytoskeleton opens the way to a better understanding of how cell muscle scaffolding is maintained through this essential protein.
Collapse
|
33
|
Schacker M, Seimetz D. From fiction to science: clinical potentials and regulatory considerations of gene editing. Clin Transl Med 2019; 8:27. [PMID: 31637541 PMCID: PMC6803602 DOI: 10.1186/s40169-019-0244-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
Gene editing technologies such as CRISPR/Cas9 have emerged as an attractive tool not only for scientific research but also for the development of medicinal products. Their ability to induce precise double strand breaks into DNA enables targeted modifications of the genome including selective knockout of genes, correction of mutations or precise insertion of new genetic material into specific loci. Gene editing-based therapies hold a great potential for the treatment of numerous diseases and the first products are already being tested in clinical trials. The treatment indications include oncological malignancies, HIV, diseases of the hematopoietic system and metabolic disorders. This article reviews ongoing preclinical and clinical studies and discusses how gene editing technologies are altering the gene therapy landscape. In addition, it focusses on the regulatory challenges associated with such therapies and how they can be tackled during the drug development process.
Collapse
Affiliation(s)
- Maria Schacker
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany.
| | - Diane Seimetz
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany
| |
Collapse
|
34
|
Wang D, Gao M, Zhang K, Jin R, Lv Y, Liu Y, Ma J, Wan Y, Gai Z, Liu Y. Molecular Genetics Analysis of 70 Chinese Families With Muscular Dystrophy Using Multiplex Ligation-Dependent Probe Amplification and Next-Generation Sequencing. Front Pharmacol 2019; 10:814. [PMID: 31404137 PMCID: PMC6669794 DOI: 10.3389/fphar.2019.00814] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Muscular dystrophy (MD) includes multiple types, of which dystrophinopathies caused by dystrophin (DMD) mutations are the most common types in children. An accurate identification of the causative mutation at the genomic level is critical for genetic counseling of the family, and analysis of genotype-phenotype correlations, as well as a reference for the development of gene therapy. Methods: Totally, 70 Chinese families with suspected MD probands were enrolled in the study. The multiplex ligation-dependent probe amplification (MLPA) was first performed to screen large deletions/duplications of DMD exons in the patients, and then, next-generation sequencing (NGS) was carried out to detect small mutations in the MLPA-negative patients. Results: Totally, 62 mutations of DMD were found in 62 probands with DMD/BMD, and two compound heterozygous mutations in LAMA2 were identified in two probands with MDC1A (a type of congenital MD), indicating that the diagnostic yield was 91.4% by MLPA plus NGS for MD diagnosis in this cohort. Out of the mutations, 51 large mutations encompassing 47 (75.8%) deletions and four duplications (6.5%) were identified by MLPA; 11 small mutations including six (9.7%) nonsense, two (3.2%) small deletions, two splice-site mutations (3.2%), and one small insertion (1.6%) were found by NGS. Large mutations were found most frequently in the hotspot region between exons 45 and 55 (70.6%). Out of the 11 patients harboring point mutations in DMD, 8 were novel mutations. Additionally, one novel mutation in LAMA2 was identified. All the novel mutations were analyzed and predicted as pathogenic according to American College of Medical Genetics and Genomics (ACMG) guideline. Finally, 34 DMD, 4 BMD, 24 BMD/DMD, and 2 MDC1A were diagnosed in the cohort. Conclusion: Our data indicated that the MLPA plus NGS can be a comprehensive and effective tool for precision diagnosis and potential treatment of MD and is particularly necessary for the patients at very young age with only two clinical indicators (persistent hyperCKemia and typical myopathy performance on electromyogram) but no definite clinical manifestations.
Collapse
Affiliation(s)
- Dong Wang
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Min Gao
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Kaihui Zhang
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Ruifeng Jin
- Neurology Department, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Yuqiang Lv
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Yong Liu
- Neurology Department, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Jian Ma
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Ya Wan
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Zhongtao Gai
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| | - Yi Liu
- Pediatric Research Institute, Qilu Children’s Hospital, Shandong University, Ji’nan, China
| |
Collapse
|
35
|
Sek AC, Moore IN, Smelkinson MG, Pak K, Minai M, Smith R, Ma M, Percopo CM, Rosenberg HF. Eosinophils Do Not Drive Acute Muscle Pathology in the mdx Mouse Model of Duchenne Muscular Dystrophy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:476-484. [PMID: 31142604 PMCID: PMC6615969 DOI: 10.4049/jimmunol.1900307] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/07/2019] [Indexed: 12/30/2022]
Abstract
Eosinophils are present in muscle lesions associated with Duchenne muscular dystrophy and dystrophin-deficient mdx mice that phenocopy this disorder. Although it has been hypothesized that eosinophils promote characteristic inflammatory muscle damage, this has not been fully examined. In this study, we generated mice with the dystrophin mutation introduced into PHIL, a strain with a transgene that directs lineage-specific eosinophil ablation. We also explored the impact of eosinophil overabundance on dystrophinopathy by introducing the dystrophin mutation into IL-5 transgenic mice. We evaluated the degree of eosinophil infiltration in association with myofiber size distribution, centralized nuclei, serum creatine kinase, and quantitative histopathology scores. Among our findings, eosinophils were prominent in the quadriceps muscles of 4-wk-old male mdx mice but no profound differences were observed in the quantitative measures of muscle damage when comparing mdx versus mdx.PHIL versus mdx.IL5tg mice, despite dramatic differences in eosinophil infiltration (CD45+CD11c-Gr1-MHC class IIloSiglecF+ eosinophils at 1.2 ± 0.34% versus <0.1% versus 20 ± 7.6% of total cells, respectively). Further evaluation revealed elevated levels of eosinophil chemoatttractants eotaxin-1 and RANTES in the muscle tissue of all three dystrophin-deficient strains; eotaxin-1 concentration in muscle correlated inversely with age. Cytokines IL-4 and IL-1R antagonist were also detected in association with eosinophils in muscle. Taken together, our findings challenge the long-held perception of eosinophils as cytotoxic in dystrophin-deficient muscle; we show clearly that eosinophil infiltration is not a driving force behind acute muscle damage in the mdx mouse strain. Ongoing studies will focus on the functional properties of eosinophils in this unique microenvironment.
Collapse
Affiliation(s)
- Albert C Sek
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- Molecular, Cellular and Integrative Physiology Program, University of California at Los Angeles, Los Angeles, CA 90095
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Margery G Smelkinson
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Katherine Pak
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Roberta Smith
- Histotechnology/Pathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Michelle Ma
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Caroline M Percopo
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Helene F Rosenberg
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
36
|
Liu C, Li L, Ge M, Gu L, Wang M, Zhang K, Su Y, Zhang Y, Liu C, Lan M, Yu Y, Wang T, Li Q, Zhao Y, Yu Z, Li N, Meng Q. Overexpression of miR-29 Leads to Myopathy that Resemble Pathology of Ullrich Congenital Muscular Dystrophy. Cells 2019; 8:cells8050459. [PMID: 31096686 PMCID: PMC6562860 DOI: 10.3390/cells8050459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022] Open
Abstract
Ullrich congenital muscular dystrophy (UCMD) bring heavy burden to patients’ families and society. Because the incidence of this disease is very low, studies in patients are extremely limited. Animal models of this disease are indispensable. UCMD belongs to extracellular matrix-related diseases. However, the disease models constructed by knocking out some pathogenic genes of human, such as the Col6a1, Col6a2, or Col6a3 gene, of mice could not mimic UCMD. The purpose of this study is to construct a mouse model which can resemble the pathology of UCMD. miR-29 is closely related to extracellular matrix deposition of tissues and organs. To address this issue, we developed a mouse model for overexpression miR-29 using Tet-on system. In the muscle-specific miR-29ab1 cluster transgenic mice model, we found that mice exhibited dyskinesia, dyspnea, and spinal anomaly. The skeletal muscle was damaged and regenerated. At the same time, we clarify the molecular mechanism of the role of miR-29 in this process. Different from human, Col4a1 and Col4a2, target genes of miR-29, are the key pathogenic genes associating with these phenotypes. This mouse model simulates the human clinical and pathological characteristics of UCMD patients and is helpful for the subsequent research and treatment of UCMD.
Collapse
Affiliation(s)
- Chuncheng Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
- The Institute of Bioengineering and Technology, Inner Mongolia University of Science and Technology, Baotou 014010, China.
| | - Lei Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Mengxu Ge
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Lijie Gu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Meng Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Kuo Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yang Su
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yuying Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Chang Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Miaomiao Lan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yingying Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Tongtong Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Qiuyan Li
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Yaofeng Zhao
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Zhengquan Yu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Ning Li
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| | - Qingyong Meng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Biological Science, China Agricultural University, Beijing 100193, China.
- The State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
37
|
Magrath P, Maforo N, Renella P, Nelson SF, Halnon N, Ennis DB. Cardiac MRI biomarkers for Duchenne muscular dystrophy. Biomark Med 2018; 12:1271-1289. [PMID: 30499689 PMCID: PMC6462870 DOI: 10.2217/bmm-2018-0125] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal inherited genetic disorder that results in progressive muscle weakness and ultimately loss of ambulation, respiratory failure and heart failure. Cardiac MRI (MRI) plays an increasingly important role in the diagnosis and clinical care of boys with DMD and associated cardiomyopathies. Conventional cardiac MRI biomarkers permit measurements of global cardiac function and presence of fibrosis, but changes in these measures are late manifestations. Emerging MRI biomarkers of myocardial function and structure include the estimation of rotational mechanics and regional strain using MRI tagging; T1-mapping; and T2-mapping, a marker of inflammation, edema and fat. These emerging biomarkers provide earlier insights into cardiac involvement in DMD, improving patient care and aiding the evaluation of emerging therapies.
Collapse
Affiliation(s)
- Patrick Magrath
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Nyasha Maforo
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Physics & Biology in Medicine IDP, University of California, Los Angeles, CA 90095, USA
| | - Pierangelo Renella
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Medicine, Division of Pediatric Cardiology, CHOC Children's Hospital, Orange, CA 92868, USA
| | - Stanley F Nelson
- Center for Duchenne Muscular Dystrophy, Department of Human Genetics, University of California, Los Angeles, CA 90095, USA
| | - Nancy Halnon
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Medicine, Division of Pediatric Cardiology, University of California, Los Angeles, CA 90024, USA
| | - Daniel B Ennis
- Department of Radiological Sciences, University of California, Los Angeles, CA 90024, USA.,Department of Bioengineering, University of California, Los Angeles, CA 90095, USA.,Physics & Biology in Medicine IDP, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
38
|
Xu R, Jia Y, Zygmunt DA, Cramer ML, Crowe KE, Shao G, Maki AE, Guggenheim HN, Hood BC, Griffin DA, Peterson E, Bolon B, Cheatham JP, Cheatham SL, Flanigan KM, Rodino-Klapac LR, Chicoine LG, Martin PT. An Isolated Limb Infusion Method Allows for Broad Distribution of rAAVrh74.MCK. GALGT2 to Leg Skeletal Muscles in the Rhesus Macaque. Mol Ther Methods Clin Dev 2018; 10:89-104. [PMID: 30073180 PMCID: PMC6070685 DOI: 10.1016/j.omtm.2018.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/05/2018] [Indexed: 10/28/2022]
Abstract
Recombinant adeno-associated virus (rAAV)rh74.MCK.GALGT2 is a muscle-specific gene therapy that is being developed to treat forms of muscular dystrophy. Here we report on an isolated limb infusion technique in a non-human primate model, where hindlimb blood flow is transiently isolated using balloon catheters to concentrate vector in targeted leg muscles. A bilateral dose of 2.5 × 1013 vector genomes (vg)/kg/limb was sufficient to induce GALGT2-induced glycosylation in 10%-60% of skeletal myofibers in all leg muscles examined. There was a 19-fold ± 6-fold average limb-wide increase in vector genomes per microgram genomic DNA at a bilateral dose of 2.5 × 1013 vg/kg/limb compared with a bilateral dose of 6 × 1012 vg/kg/limb. A unilateral dose of 6 × 1013 vg/kg/limb showed a 12- ± 3-fold increase in treated limb muscles compared to contralateral untreated limb muscles, which received vector only after release into the systemic circulation from the treated limb. Variability in AAV biodistribution between different segments of the same muscle was 125% ± 18% for any given dose, while variability between the same muscle for any given treatment dose was 45% ± 7%. These experiments demonstrate that treatment of muscles throughout the leg with rAAVrh74.MCK.GALGT2 can be accomplished safely using an isolated limb infusion technique, where balloon catheters transiently isolate the limb vasculature, but that intra- and inter-muscle transduction variability is a significant issue.
Collapse
Affiliation(s)
- Rui Xu
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Ying Jia
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Deborah A. Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Megan L. Cramer
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Kelly E. Crowe
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Guohong Shao
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Agatha E. Maki
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Haley N. Guggenheim
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Benjamin C. Hood
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Danielle A. Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | - Ellyn Peterson
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
| | | | - John P. Cheatham
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Sharon L. Cheatham
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Kevin M. Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Louise R. Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Louis G. Chicoine
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Paul T. Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| |
Collapse
|
39
|
Crowe KE, Shao G, Flanigan KM, Martin PT. N-terminal α Dystroglycan (αDG-N): A Potential Serum Biomarker for Duchenne Muscular Dystrophy. J Neuromuscul Dis 2018; 3:247-260. [PMID: 27854211 DOI: 10.3233/jnd-150127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a severe, progressive, neuromuscular disorder of childhood. While a number of serum factors have been identified as potential biomarkers of DMD, none, as yet, are proteins within the dystrophin-associated glycoprotein (DAG) complex. OBJECTIVE We have developed an immobilized serum ELISA assay to measure the expression of a constitutively cleaved and secreted component of the DAG complex, the N-terminal domain of α dystroglycan (αDG-N), and assayed relative expression in serum from muscular dystrophy patients and normal controls. METHODS ELISAs of immobilized patient or mouse serum and Western blots were used to assess αDG-N expression. RESULTS Immobilization of diluted serum on ELISA plates was important for this assay, as methods to measure serum αDG-N in solution were less robust. αDG-N ELISA signals were significantly reduced in DMD serum (27±3% decrease, n = 9, p < 0.001) relative to serum from otherwise normal controls (n = 38), and calculated serum αDG-N concentrations were reduced in DMD relative to normal (p < 0.01) and Becker Muscular Dystrophy (n = 11, p < 0.05) patient serum. By contrast, ELISA signals from patients with Inclusion Body Myositis were not different than normal (4±3% decrease, n = 8, p = 0.99). αDG-N serum signals were also significantly reduced in utrophin-deficient mdx mice as compared to mdx and wild type mice. CONCLUSIONS Our results are the first demonstration of a component of the DAG complex as a potential serum biomarker in DMD. Such a serum measure could be further developed as a tool to help reflect overall muscle DAG complex expression or stability.
Collapse
Affiliation(s)
- Kelly E Crowe
- Graduate Program in Molecular Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Guohong Shao
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
40
|
Clinical Utility Gene Card for: Becker muscular dystrophy. Eur J Hum Genet 2018; 26:1065-1071. [PMID: 29467387 DOI: 10.1038/s41431-017-0064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 09/10/2017] [Accepted: 11/23/2017] [Indexed: 12/28/2022] Open
|
41
|
Next Generation Sequencing approach to molecular diagnosis of Duchenne muscular dystrophy; identification of a novel mutation. Gene 2018; 644:1-3. [DOI: 10.1016/j.gene.2017.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 11/22/2017] [Accepted: 12/07/2017] [Indexed: 11/20/2022]
|
42
|
CUGC for Duchenne muscular dystrophy (DMD). Eur J Hum Genet 2018; 26:749-757. [PMID: 29330543 DOI: 10.1038/s41431-017-0013-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 08/08/2017] [Accepted: 09/09/2017] [Indexed: 11/08/2022] Open
|
43
|
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disorder that causes progressive weakness and wasting of skeletal muscular and myocardium in boys due to mutation of dystrophin. The structural integrity of each individual skeletal and cardiac myocyte is significantly compromised upon physical stress due to the absence of dystrophin. The progressive destruction of systemic musculature and myocardium causes affected patients to develop multiple organ disabilities, including loss of ambulation, physical immobility, neuromuscular scoliosis, joint contracture, restrictive lung disease, obstructive sleep apnea, and cardiomyopathy. There are some central nervous system-related medical problems, as dystrophin is also expressed in the neuronal tissues. Although principal management is to mainly delay the pathological process, an enhanced understanding of underlying pathological processes has significantly improved quality of life and longevity for DMD patients. Future research in novel molecular approach is warranted to answer unanswered questions.
Collapse
Affiliation(s)
- Takeshi Tsuda
- Nemours Cardiac Center, Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE, 19803, USA.
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Fraysse B, Barthélémy I, Qannari EM, Rouger K, Thorin C, Blot S, Le Guiner C, Chérel Y, Hogrel JY. Gait characterization in golden retriever muscular dystrophy dogs using linear discriminant analysis. BMC Musculoskelet Disord 2017; 18:153. [PMID: 28403854 PMCID: PMC5388997 DOI: 10.1186/s12891-017-1494-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/20/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accelerometric analysis of gait abnormalities in golden retriever muscular dystrophy (GRMD) dogs is of limited sensitivity, and produces highly complex data. The use of discriminant analysis may enable simpler and more sensitive evaluation of treatment benefits in this important preclinical model. METHODS Accelerometry was performed twice monthly between the ages of 2 and 12 months on 8 healthy and 20 GRMD dogs. Seven accelerometric parameters were analysed using linear discriminant analysis (LDA). Manipulation of the dependent and independent variables produced three distinct models. The ability of each model to detect gait alterations and their pattern change with age was tested using a leave-one-out cross-validation approach. RESULTS Selecting genotype (healthy or GRMD) as the dependent variable resulted in a model (Model 1) allowing a good discrimination between the gait phenotype of GRMD and healthy dogs. However, this model was not sufficiently representative of the disease progression. In Model 2, age in months was added as a supplementary dependent variable (GRMD_2 to GRMD_12 and Healthy_2 to Healthy_9.5), resulting in a high overall misclassification rate (83.2%). To improve accuracy, a third model (Model 3) was created in which age was also included as an explanatory variable. This resulted in an overall misclassification rate lower than 12%. Model 3 was evaluated using blinded data pertaining to 81 healthy and GRMD dogs. In all but one case, the model correctly matched gait phenotype to the actual genotype. Finally, we used Model 3 to reanalyse data from a previous study regarding the effects of immunosuppressive treatments on muscular dystrophy in GRMD dogs. Our model identified significant effect of immunosuppressive treatments on gait quality, corroborating the original findings, with the added advantages of direct statistical analysis with greater sensitivity and more comprehensible data representation. CONCLUSIONS Gait analysis using LDA allows for improved analysis of accelerometry data by applying a decision-making analysis approach to the evaluation of preclinical treatment benefits in GRMD dogs.
Collapse
Affiliation(s)
| | - Inès Barthélémy
- INSERM U955-E10 Biology of the NeuroMuscular System, 94000, Créteil, France.,Université Paris-Est, École Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France.,Faculté de Médecine, 94000, Créteil, France
| | - El Mostafa Qannari
- LUNAM University, ONIRIS, National College of Veterinary Medicine, Food Science, and Engineering, USC "Sensometrics and Chemometrics Laboratory", Nantes, France
| | - Karl Rouger
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Chantal Thorin
- Nutrition and Endocrinology Unit, ONIRIS, National College of Veterinary Medicine, Food Science, and Engineering, Nantes, France
| | - Stéphane Blot
- INSERM U955-E10 Biology of the NeuroMuscular System, 94000, Créteil, France.,Université Paris-Est, École Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France.,Faculté de Médecine, 94000, Créteil, France
| | | | - Yan Chérel
- Atlantic Gene Therapies, INRA UMR 703, ONIRIS, Nantes, France
| | - Jean-Yves Hogrel
- Neuromuscular Physiology and Evaluation Lab, Institute of Myology, Paris, France
| |
Collapse
|
45
|
Van Ruiten H, Bushby K, Guglieri M. State-Of-The-Art Advances in Duchenne Muscular Dystrophy. EUROPEAN MEDICAL JOURNAL 2017. [DOI: 10.33590/emj/10311993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe and fatal muscle condition affecting young children. Without interventions, affected boys lose the ability to walk independently by the age of 10 and develop progressive cardiac and respiratory failure. The last 20 years have seen a change in the natural history of DMD following improvements in clinical care and proactive interventions to manage complications of the disease. An international collaboration of DMD experts has created care imperatives for best practice in DMD; these are now available in 30 different languages and are disseminated worldwide. An update of these care recommendations is currently under review.
More recently, the field has seen encouraging scientific progress in regard to new therapeutic approaches of which a large number are currently being evaluated in clinical trials. With time, improvements in clinical care and access to new treatments and innovations are changing the natural course of DMD, from a relentless progressive illness with death in teenage years to a more chronic illness with a good quality of life and increased life expectancy. This is a particularly encouraging time for DMD, and experiences built in the muscular dystrophy field are likely to be of benefit to the development of new approaches and therapies in other rare diseases.
Collapse
Affiliation(s)
- Henriette Van Ruiten
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Katherine Bushby
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, UK
| |
Collapse
|
46
|
Rasolonjatovo B, Pitard B, Haudebourg T, Bennevault V, Guégan P. Synthesis of tetraarm star block copolymer based on polytetrahydrofuran and poly(2-methyl-2-oxazoline) for gene delivery applications. Eur Polym J 2017. [DOI: 10.1016/j.eurpolymj.2016.09.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Petryk A, Polgreen LE, Grames M, Lowe DA, Hodges JS, Karachunski P. Feasibility and tolerability of whole-body, low-intensity vibration and its effects on muscle function and bone in patients with dystrophinopathies: a pilot study. Muscle Nerve 2017; 55:875-883. [PMID: 27718512 PMCID: PMC5385164 DOI: 10.1002/mus.25431] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 10/03/2016] [Accepted: 10/06/2016] [Indexed: 12/12/2022]
Abstract
Introduction Dystrophinopathies are X‐linked muscle degenerative disorders that result in progressive muscle weakness complicated by bone loss. This study's goal was to evaluate feasibility and tolerability of whole‐body, low‐intensity vibration (WBLIV) and its potential effects on muscle and bone in patients with Duchenne or Becker muscular dystrophy. Methods This 12‐month pilot study included 5 patients (age 5.9–21.7 years) who used a low‐intensity Marodyne LivMD plate vibrating at 30–90 Hz for 10 min/day for the first 6 months. Timed motor function tests, myometry, and peripheral quantitative computed tomography were performed at baseline and at 6 and 12 months. Results Motor function and lower extremity muscle strength remained either unchanged or improved during the intervention phase, followed by deterioration after WBLIV discontinuation. Indices of bone density and geometry remained stable in the tibia. Conclusions WBLIV was well tolerated and appeared to have a stabilizing effect on lower extremity muscle function and bone measures. Muscle Nerve55: 875–883, 2017
Collapse
Affiliation(s)
- Anna Petryk
- Division of Pediatric Endocrinology, University of Minnesota Masonic Children's Hospital, 2450 Riverside Avenue, Minneapolis, Minnesota, 55454, USA.,Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lynda E Polgreen
- Division of Pediatric Endocrinology and Metabolism, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Molly Grames
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Dawn A Lowe
- Department of Physical Medicine and Rehabilitation, University of Minnesota, Minneapolis, Minnesota, USA
| | - James S Hodges
- Division of Biostatistics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter Karachunski
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA.,Department of Neurology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
48
|
Delalande O, Czogalla A, Hubert JF, Sikorski A, Le Rumeur E. Dystrophin and Spectrin, Two Highly Dissimilar Sisters of the Same Family. Subcell Biochem 2017; 82:373-403. [PMID: 28101868 DOI: 10.1007/978-3-319-49674-0_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dystrophin and Spectrin are two proteins essential for the organization of the cytoskeleton and for the stabilization of membrane cells. The comparison of these two sister proteins, and with the dystrophin homologue utrophin, enables us to emphasise that, despite a similar topology with common subdomains and a common structural basis of a three-helix coiled-coil, they show a large range of dissimilarities in terms of genetics, cell expression and higher level structural organisation. Interactions with cellular partners, including proteins and membrane phospholipids, also show both strikingly similar and very different behaviours. The differences between dystrophin and spectrin are also illustrated by the large variety of pathological anomalies emerging from the dysfunction or the absence of these proteins, showing that they are keystones in their function of providing a scaffold that sustains cell structure.
Collapse
Affiliation(s)
- Olivier Delalande
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France.
| | - Aleksander Czogalla
- Biotechnology Faculty, Department of Cytobiochemistry, University of Wrocław, ul. joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Jean-François Hubert
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France
| | - Aleksander Sikorski
- Biotechnology Faculty, Department of Cytobiochemistry, University of Wrocław, ul. joliot-Curie 14a, 50-383, Wroclaw, Poland
| | - Elisabeth Le Rumeur
- Institut de Génétique et Développement de Rennes, UMR CNRS 6290, Université de Rennes 1, Rennes, France
| |
Collapse
|
49
|
Zygmunt DA, Crowe KE, Flanigan KM, Martin PT. Comparison of Serum rAAV Serotype-Specific Antibodies in Patients with Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, Inclusion Body Myositis, or GNE Myopathy. Hum Gene Ther 2016; 28:737-746. [PMID: 28042944 DOI: 10.1089/hum.2016.141] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Recombinant adeno-associated virus (rAAV) is a commonly used gene therapy vector for the delivery of therapeutic transgenes in a variety of human diseases, but pre-existing serum antibodies to viral capsid proteins can greatly inhibit rAAV transduction of tissues. Serum was assayed from patients with Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), inclusion body myositis (IBM), and GNE myopathy (GNE). These were compared to serum from otherwise normal human subjects to determine the extent of pre-existing serum antibodies to rAAVrh74, rAAV1, rAAV2, rAAV6, rAAV8, and rAAV9. In almost all cases, patients with measurable titers to one rAAV serotype showed titers to all other serotypes tested, with average titers to rAAV2 being highest in all instances. Twenty-six percent of all young normal subjects (<18 years old) had measurable rAAV titers to all serotypes tested, and this percentage increased to almost 50% in adult normal subjects (>18 years old). Fifty percent of all IBM and GNE patients also had antibody titers to all rAAV serotypes, while only 18% of DMD and 0% of BMD patients did. In addition, serum-naïve macaques treated systemically with rAAVrh74 could develop cross-reactive antibodies to all other serotypes tested at 24 weeks post treatment. These data demonstrate that most DMD and BMD patients should be amenable to vascular rAAV-mediated treatment without the concern of treatment blockage by pre-existing serum rAAV antibodies, and that serum antibodies to rAAVrh74 are no more common than those for rAAV6, rAAV8, or rAAV9.
Collapse
Affiliation(s)
- Deborah A Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University , Columbus, Ohio
| | - Kelly E Crowe
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University , Columbus, Ohio
| | - Kevin M Flanigan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University , Columbus, Ohio
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University , Columbus, Ohio
| |
Collapse
|
50
|
Tjondrokoesoemo A, Schips TG, Sargent MA, Vanhoutte D, Kanisicak O, Prasad V, Lin SCJ, Maillet M, Molkentin JD. Cathepsin S Contributes to the Pathogenesis of Muscular Dystrophy in Mice. J Biol Chem 2016; 291:9920-8. [PMID: 26966179 DOI: 10.1074/jbc.m116.719054] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 11/06/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and β-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jeffery D Molkentin
- From the Department of Pediatrics and Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229
| |
Collapse
|