1
|
Matesanz SE, Edelson JB, Iacobellis KA, Mejia E, Brandsema JF, Wittlieb-Weber CA, Okunowo O, Griffis H, Lin KY. Subspecialty Health Care Utilization in Pediatric Patients With Muscular Dystrophy in the United States. Neurol Clin Pract 2024; 14:e200312. [PMID: 38855715 PMCID: PMC11160481 DOI: 10.1212/cpj.0000000000200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/25/2024] [Indexed: 06/11/2024]
Abstract
Background and Objectives Standards of care exist to optimize outcomes in Duchenne and Becker muscular dystrophy (DBMD), caused by alterations in the DMD gene; however, there are limited data regarding health care access in these patients. This study aims to characterize outpatient subspecialty care utilization in pediatric patients with DBMD. Methods This retrospective cohort study used administrative claims data from IBM MarketScan Medicaid and Commercial Claims and Encounters Research Databases (2013-2018). Male patients 1-18 years with an ICD-9/10 diagnosis code for hereditary progressive muscular dystrophy between January 1, 2013, and December 31, 2017, were included. Participants were stratified into 3 age cohorts: 1-6 years, 7-12 years, and 13-18 years. The primary outcome was rate of annual neurology visits. Secondary outcomes included annual follow-up rates in other subspecialties and proportion of days covered (PDC) by corticosteroids. Results A total of 1,386 patients met inclusion-347 (25.0%) age 1-6 years, 502 (36.2%) age 7-12 years, and 537 (38.7%) age 13-18 years. Heart failure, respiratory failure, and technology dependence increased with age (p for all<0.05). The rate of neurology visits per person-year was 0.36 and did not differ by age. Corticosteroid use was low; 30% of person-years (1452/4829) had a PDC ≥20%. Medicaid insurance was independently associated with a lower likelihood of annual neurology follow-up (OR 0.23; 95% CI 0.18-0.28). Discussion The rate of annual neurology follow-up and corticosteroid use in patients with DBMD is low. Medicaid insurance status was independently associated with a decreased likelihood of neurology follow-up, while age was not, suggesting that factors other than disease severity influence neurology care access. Identifying barriers to regular follow-up is critical in improving outcomes for patients with DBMD.
Collapse
Affiliation(s)
- Susan E Matesanz
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Jonathan B Edelson
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Katherine A Iacobellis
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Erika Mejia
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - John F Brandsema
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Carol A Wittlieb-Weber
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Oluwatimilehin Okunowo
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Heather Griffis
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| | - Kimberly Y Lin
- Division of Neurology (SEM, JFB); Division of Cardiology (JBE, KAI, EM, CAW-W, KYL), Cardiac Center, the Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine; Leonard Davis Institute Center for Healthcare Economics (JBE); Cardiovascular Outcomes, Quality, and Evaluative Research Center (JBE), University of Pennsylvania, Philadelphia; and Data Science and Biostatistics Unit (OO, HG), Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia
| |
Collapse
|
2
|
Bhattarai P, Karki M. Recurrent Spontaneous Pneumothorax in a Young Male With Duchenne Muscular Dystrophy Following COVID-19 Infection. Cureus 2024; 16:e52408. [PMID: 38371092 PMCID: PMC10869316 DOI: 10.7759/cureus.52408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 02/20/2024] Open
Abstract
Duchene muscular dystrophy (DMD) is a genetic disorder primarily affecting males. It is characterized by progressive muscle tissue degeneration. Patients with DMD are at an increased risk of respiratory infections, including coronavirus disease 20019 (COVID-19), due to weakened respiratory muscles. We present a case of a young male with DMD who experienced recurrent pneumothorax 10 months after recovering from a COVID-19 infection. The patient required prompt medical intervention, including a chest tube, multiple surgeries, and mechanical pleurodesis. This case highlights the importance of recognizing recurring pneumothorax as a potential complication of COVID-19, particularly in patients with underlying neuromuscular disorders, as it is a medical emergency requiring prompt treatment to prevent respiratory compromise.
Collapse
Affiliation(s)
- Pramod Bhattarai
- Pulmonary Medicine, Howard University Hospital, Washington, DC, USA
- Critical Care Medicine, Larkin Community Hospital Palm Springs Campus, Hialeah, USA
- Internal Medicine, Harlem Hospital Center, New York, USA
| | - Monika Karki
- Internal Medicine, The Brooklyn Hospital Center, Brooklyn, USA
| |
Collapse
|
3
|
Soni N, Nandi G, Chaudhary M, Bissa B. The role of ncRNA in the co-regulation of autophagy and exosome pathways during cancer progression. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119523. [PMID: 37348764 DOI: 10.1016/j.bbamcr.2023.119523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
Since its discovery a few decades ago, autophagy has been recognized as a crucial signaling pathway, linked to the recycling of cellular components in nutrient stress. Autophagy is a two-way sword, playing a dual role in tumorigenesis. In this catabolic process, dysfunctional organelles, biomolecules, and misfolded proteins are sequestered in the autophagosome and sent to the lysosome for degradation. Alongside, there are cellular messengers called exosomes, which are released from cells and are known to communicate and regulate metabolism in recipient cells. Multivesicular bodies (MVB) act as the intricate link between autophagy and exosome pathways. The continuous crosstalk between the two pathways is coordinated and regulated by multiple players among which ncRNA is the emerging candidates. The exosomes carry varied cargo of which non-coding RNA exerts an immediate regulatory effect on recipient cells. ncRNA is known to exhibit dual behavior in both promoting and inhibiting tumor growth. There is increasing evidence for the involvement of ncRNAs' in the regulation of different hallmarks of cancer. Different ncRNAs are involved in the co-regulation of autophagy and exosome pathways and therefore represent a superior therapeutic approach to target cancer chemoresistance. Here, we will discuss the ncRNA involved in regulating autophagy, and exosomes pathways and its relevance in cancer therapeutics.
Collapse
Affiliation(s)
- Naveen Soni
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Gargi Nandi
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Megha Chaudhary
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Bhawana Bissa
- Dept. of Biochemistry, Central University of Rajasthan, Ajmer, Rajasthan, India.
| |
Collapse
|
4
|
Wei Y, Su Q, Li X. Identification of hub genes related to Duchenne muscular dystrophy by weighted gene co-expression network analysis. Medicine (Baltimore) 2022; 101:e32603. [PMID: 36596079 PMCID: PMC9803489 DOI: 10.1097/md.0000000000032603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The study was aimed to analyze the potential gene modules and hub genes of Duchenne muscular dystrophy (DMD) by weighted gene co-expression network analysis. METHODS Based on the muscular dystrophy tissue expression profiling microarray GSE13608 from gene expression omnibus, gene co-expression modules were analyzed using weighted gene co-expression network analysis, gene modules related to DMD were screened, gene ontology and Kyoto encyclopedia of genes and genomes enrichment analyses were performed, and signature genes in the modules were screened. The protein-protein interaction network was constructed through Cytoscape, and hub genes were identified. The expression of hub genes in DMD versus normal muscle tissue was calculated in GSE6011. RESULTS 12 co-expressed gene modules were identified, among which black module was significantly related to DMD. The characteristic genes in the module were enriched in the regulation of immune effector processes, immune response mediated by immunoglobulin, immune response mediated by B cells, etc. SERPING1, F13A1, C1S, C1R, and HLA-DPA1 were considered as hub genes in protein-protein interaction network. Analysis of GSE6011 shows that expression of SERPING1, F13A1, C1S, C1R, and HLA-DPA1 in tissues of DMD patients were higher than normal. CONCLUSION SERPING1, F13A1, C1S, C1R, and HLA-DPA1 may participate in the development of DMD by regulating innate immunity and inflammation, and they are expected to be a potential biomarker and novel therapeutic targets for DMD.
Collapse
Affiliation(s)
- Yanning Wei
- School of Public Health, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Qisheng Su
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaohong Li
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
- * Correspondence: Xiaohong Li, Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China (e-mail: )
| |
Collapse
|
5
|
Sherlock SP, Palmer J, Wagner KR, Abdel-Hamid HZ, Bertini E, Tian C, Mah JK, Kostera-Pruszczyk A, Muntoni F, Guglieri M, Brandsema JF, Mercuri E, Butterfield RJ, McDonald CM, Charnas L, Marraffino S. Quantitative magnetic resonance imaging measures as biomarkers of disease progression in boys with Duchenne muscular dystrophy: a phase 2 trial of domagrozumab. J Neurol 2022; 269:4421-4435. [PMID: 35396602 PMCID: PMC9294028 DOI: 10.1007/s00415-022-11084-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/11/2022] [Accepted: 03/12/2022] [Indexed: 01/14/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive, neuromuscular disorder caused by mutations in the DMD gene that results in a lack of functional dystrophin protein. Herein, we report the use of quantitative magnetic resonance imaging (MRI) measures as biomarkers in the context of a multicenter phase 2, randomized, placebo-controlled clinical trial evaluating the myostatin inhibitor domagrozumab in ambulatory boys with DMD (n = 120 aged 6 to < 16 years). MRI scans of the thigh to measure muscle volume, muscle volume index (MVI), fat fraction, and T2 relaxation time were obtained at baseline and at weeks 17, 33, 49, and 97 as per protocol. These quantitative MRI measurements appeared to be sensitive and objective biomarkers for evaluating disease progression, with significant changes observed in muscle volume, MVI, and T2 mapping measures over time. To further explore the utility of quantitative MRI measures as biomarkers to inform longer term functional changes in this cohort, a regression analysis was performed and demonstrated that muscle volume, MVI, T2 mapping measures, and fat fraction assessment were significantly correlated with longer term changes in four-stair climb times and North Star Ambulatory Assessment functional scores. Finally, less favorable baseline measures of MVI, fat fraction of the muscle bundle, and fat fraction of lean muscle were significant risk factors for loss of ambulation over a 2-year monitoring period. These analyses suggest that MRI can be a valuable tool for use in clinical trials and may help inform future functional changes in DMD.Trial registration: ClinicalTrials.gov identifier, NCT02310763; registered December 2014.
Collapse
Affiliation(s)
| | | | - Kathryn R Wagner
- Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hoda Z Abdel-Hamid
- Division of Child Neurology, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Enrico Bertini
- Unit of Neuromuscular Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Cuixia Tian
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Jean K Mah
- Alberta Children's Hospital, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle, UK
| | | | - Eugenio Mercuri
- Pediatric Neurology, Catholic University, Rome, Italy
- Centro Nemo, Fondazione Policlinico Gemelli IRCCS, Rome, Italy
| | | | | | | | | |
Collapse
|
6
|
Song MH, Yoo J, Oh JG, Kook H, Park WJ, Jeong D. Matricellular Protein CCN5 Gene Transfer Ameliorates Cardiac and Skeletal Dysfunction in mdx/utrn (±) Haploinsufficient Mice by Reducing Fibrosis and Upregulating Utrophin Expression. Front Cardiovasc Med 2022; 9:763544. [PMID: 35557546 PMCID: PMC9088811 DOI: 10.3389/fcvm.2022.763544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/31/2022] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disorder characterized by progressive muscle degeneration due to dystrophin gene mutations. Patients with DMD initially experience muscle weakness in their limbs during adolescence. With age, patients develop fatal respiratory and cardiac dysfunctions. During the later stages of the disease, severe cardiac fibrosis occurs, compromising cardiac function. Previously, our research showed that the matricellular protein CCN5 has antifibrotic properties. Therefore, we hypothesized that CCN5 gene transfer would ameliorate cardiac fibrosis and thus improve cardiac function in DMD-induced cardiomyopathy. We utilized mdx/utrn (±) haploinsufficient mice that recapitulated the DMD-disease phenotypes and used an adeno-associated virus serotype-9 viral vector for CCN5 gene transfer. We evaluated the onset of cardiac dysfunction using echocardiography and determined the experimental starting point in 13-month-old mice. Two months after CCN5 gene transfer, cardiac function was significantly enhanced, and cardiac fibrosis was ameliorated. Additionally, running performance was improved in CCN5 gene-transfected mice. Furthermore, in silico gene profiling analysis identified utrophin as a novel transcriptional target of CCN5. This was supplemented by a utrophin promoter assay and RNA-seq analysis, which confirmed that CCN5 was directly associated with utrophin expression. Our results showed that CCN5 may be a promising therapeutic molecule for DMD-induced cardiac and skeletal dysfunction.
Collapse
Affiliation(s)
- Min Ho Song
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Jimeen Yoo
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jae Gyun Oh
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Hyun Kook
- Basic Research Laboratory, Chonnam National University Medical School, Gwangju, South Korea
| | - Woo Jin Park
- College of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Dongtak Jeong
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Molecular and Life Science, College of Science and Convergence Technology, Hanyang University-ERICA, Ansan, South Korea
| |
Collapse
|
7
|
Lingineni K, Aggarwal V, Morales JF, Conrado DJ, Corey D, Vong C, Burton J, Larkindale J, Romero K, Schmidt S, Kim S. Development of a model-based clinical trial simulation platform to optimize the design of clinical trials for Duchenne muscular dystrophy. CPT Pharmacometrics Syst Pharmacol 2022; 11:318-332. [PMID: 34877803 PMCID: PMC8923721 DOI: 10.1002/psp4.12753] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022] Open
Abstract
Early clinical trials of therapies to treat Duchenne muscular dystrophy (DMD), a fatal genetic X‐linked pediatric disease, have been designed based on the limited understanding of natural disease progression and variability in clinical measures over different stages of the continuum of the disease. The objective was to inform the design of DMD clinical trials by developing a disease progression model‐based clinical trial simulation (CTS) platform based on measures commonly used in DMD trials. Data were integrated from past studies through the Duchenne Regulatory Science Consortium founded by the Critical Path Institute (15 clinical trials and studies, 1505 subjects, 27,252 observations). Using a nonlinear mixed‐effects modeling approach, longitudinal dynamics of five measures were modeled (NorthStar Ambulatory Assessment, forced vital capacity, and the velocities of the following three timed functional tests: time to stand from supine, time to climb 4 stairs, and 10 meter walk‐run time). The models were validated on external data sets and captured longitudinal changes in the five measures well, including both early disease when function improves as a result of growth and development and the decline in function in later stages. The models can be used in the CTS platform to perform trial simulations to optimize the selection of inclusion/exclusion criteria, selection of measures, and other trial parameters. The data sets and models have been reviewed by the US Food and Drug Administration and the European Medicines Agency; have been accepted into the Fit‐for‐Purpose and Qualification for Novel Methodologies pathways, respectively; and will be submitted for potential endorsement by both agencies.
Collapse
Affiliation(s)
- Karthik Lingineni
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Diane Corey
- Critical Path Institute, Tucson, Arizona, USA
| | - Camille Vong
- Global Product Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | | | | | | | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | |
Collapse
|
8
|
Wharton's Jelly-Derived Mesenchymal Stem Cells Reduce Fibrosis in a Mouse Model of Duchenne Muscular Dystrophy by Upregulating microRNA 499. Biomedicines 2021; 9:biomedicines9091089. [PMID: 34572277 PMCID: PMC8469349 DOI: 10.3390/biomedicines9091089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/23/2021] [Indexed: 01/10/2023] Open
Abstract
The aim of this study was to evaluate the therapeutic effects and mechanisms of Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) in an animal model of Duchenne muscular dystrophy (DMD). Mdx mice (3-5 months old) were administered five different doses of WJ-MSCs through their tail veins. A week after injection, grip strength measurements, creatine kinase (CK) assays, immunohistochemistry, and western blots were performed for comparison between healthy mice, mdx control mice, and WJ-MSC-injected mdx mice. WJ-MSCs exerted dose-dependent multisystem therapeutic effects in mdx mice, by decreasing CK, recovering normal behavior, regenerating muscle, and reducing apoptosis and fibrosis in skeletal muscle. We also confirmed that miR-499-5p is significantly downregulated in mdx mice, and that intravenous injection of WJ-MSCs enhanced its expression, leading to anti-fibrotic effects via targeting TGFβR 1 and 3. Thus, WJ-MSCs may represent novel allogeneic "off-the-shelf" cellular products for the treatment of DMD and possibly other muscle disorders.
Collapse
|
9
|
Luan P, D'Amico D, Andreux PA, Laurila PP, Wohlwend M, Li H, Imamura de Lima T, Place N, Rinsch C, Zanou N, Auwerx J. Urolithin A improves muscle function by inducing mitophagy in muscular dystrophy. Sci Transl Med 2021; 13:13/588/eabb0319. [PMID: 33827972 DOI: 10.1126/scitranslmed.abb0319] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 11/25/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy, and despite advances in genetic and pharmacological disease-modifying treatments, its management remains a major challenge. Mitochondrial dysfunction contributes to DMD, yet the mechanisms by which this occurs remain elusive. Our data in experimental models and patients with DMD show that reduced expression of genes involved in mitochondrial autophagy, or mitophagy, contributes to mitochondrial dysfunction. Mitophagy markers were reduced in skeletal muscle and in muscle stem cells (MuSCs) of a mouse model of DMD. Administration of the mitophagy activator urolithin A (UA) rescued mitophagy in DMD worms and mice and in primary myoblasts from patients with DMD, increased skeletal muscle respiratory capacity, and improved MuSCs' regenerative ability, resulting in the recovery of muscle function and increased survival in DMD mouse models. These data indicate that restoration of mitophagy alleviates symptoms of DMD and suggest that UA may have potential therapeutic applications for muscular dystrophies.
Collapse
Affiliation(s)
- Peiling Luan
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Davide D'Amico
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.,Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pénélope A Andreux
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Pirkka-Pekka Laurila
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Martin Wohlwend
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Hao Li
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Tanes Imamura de Lima
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Nicolas Place
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, Ecole Polytechnique Fédérale de Lausanne (EPFL) Innovation Park, 1015 Lausanne, Switzerland
| | - Nadège Zanou
- Institute of Sport Sciences, Quartier UNIL-Centre, Faculty of Biology-Medicine, University of Lausanne, Bâtiment Synathlon, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
10
|
Luce L, Carcione M, Mazzanti C, Buonfiglio PI, Dalamón V, Mesa L, Dubrovsky A, Corderí J, Giliberto F. Theragnosis for Duchenne Muscular Dystrophy. Front Pharmacol 2021; 12:648390. [PMID: 34149409 PMCID: PMC8209366 DOI: 10.3389/fphar.2021.648390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/12/2022] Open
Abstract
Dystrophinopathies cover a spectrum of rare progressive X-linked muscle diseases, arising from DMD mutations. They are among the most common pediatric muscular dystrophies, being Duchenne muscular dystrophy (DMD) the most severe form. Despite the fact that there is still no cure for these serious diseases, unprecedented advances are being made for the development of therapies for DMD. Some of which are already conditionally approved: exon skipping and premature stop codon read-through. The present work aimed to characterize the mutational spectrum of DMD in an Argentinian cohort, to identify candidates for available pharmacogenetic treatments and finally, to conduct a comparative analysis of the Latin American (LA) frequencies of mutations amenable for available DMD therapies. We studied 400 patients with clinical diagnosis of dystrophinopathy, implementing a diagnostic molecular algorithm including: MLPA/PCR/Sanger/Exome and bioinformatics. We also performed a meta-analysis of LA's metrics for DMD available therapies. The employed algorithm resulted effective for the achievement of differential diagnosis, reaching a detection rate of 97%. Because of this, corticosteroid treatment was correctly indicated and validated in 371 patients with genetic confirmation of dystrophinopathy. Also, 20 were eligible for exon skipping of exon 51, 21 for exon 53, 12 for exon 45 and another 70 for premature stop codon read-through therapy. We determined that 87.5% of DMD patients will restore the reading frame with the skipping of only one exon. Regarding nonsense variants, UGA turned out to be the most frequent premature stop codon observed (47%). According to the meta-analysis, only four LA countries (Argentina, Brazil, Colombia and Mexico) provide the complete molecular algorithm for dystrophinopathies. We observed different relations among the available targets for exon skipping in the analyzed populations, but a more even proportion of nonsense variants (∼40%). In conclusion, this manuscript describes the theragnosis carried out in Argentinian dystrophinopathy patients. The implemented molecular algorithm proved to be efficient for the achievement of differential diagnosis, which plays a crucial role in patient management, determination of the standard of care and genetic counseling. Finally, this work contributes with the international efforts to characterize the frequencies and variants in LA, pillars of drug development and theragnosis.
Collapse
Affiliation(s)
- Leonela Luce
- Laboratorio de Distrofinopatías, Cátedra de Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Micaela Carcione
- Laboratorio de Distrofinopatías, Cátedra de Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Chiara Mazzanti
- Laboratorio de Distrofinopatías, Cátedra de Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paula I Buonfiglio
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) "Dr. Héctor N. Torres", CONICET, Buenos Aires, Argentina
| | - Viviana Dalamón
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (INGEBI) "Dr. Héctor N. Torres", CONICET, Buenos Aires, Argentina
| | - Lilia Mesa
- Instituto de Neurociencias, Fundación Favaloro, Buenos Aires, Argentina
| | - Alberto Dubrovsky
- Instituto de Neurociencias, Fundación Favaloro, Buenos Aires, Argentina
| | - José Corderí
- Instituto de Neurociencias, Fundación Favaloro, Buenos Aires, Argentina
| | - Florencia Giliberto
- Laboratorio de Distrofinopatías, Cátedra de Genética, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Inmunología, Genética y Metabolismo (INIGEM), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
11
|
A Phase 1/2 Study of Flavocoxid, an Oral NF-κB Inhibitor, in Duchenne Muscular Dystrophy. Brain Sci 2021; 11:brainsci11010115. [PMID: 33467104 PMCID: PMC7830560 DOI: 10.3390/brainsci11010115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/15/2022] Open
Abstract
Flavocoxid is a blended extract containing baicalin and catechin with potent antioxidant and anti-inflammatory properties due to the inhibition of the cyclooxygenase (COX) and 5-lipoxygenase (5-LOX) enzymes, nuclear factor-κB (NF-κB), tumor necrosis factor (TNF)-alpha, and the mitogen-activated protein kinases (MAPKs) pathways. This phase 1/2 study was designed to assess the safety and tolerability of flavocoxid in patients with Duchenne muscular dystrophy (DMD). Thirty-four patients were recruited: 17 were treated with flavocoxid at an oral dose of 250 or 500 mg, according to body weight, for one year; 17 did not receive flavocoxid and served as controls. The treatment was well tolerated and nobody dropped out. Flavocoxid induced a significant reduction in serum interleukin (IL)-1 beta and TNF-alpha only in the group of DMD boys on add-on therapy (flavocoxid added to steroids for at least six months). The decrease in IL-1 beta was higher in younger boys. The serum H2O2 concentrations significantly decreased in patients treated with flavocoxid alone with a secondary reduction of serum glutathione peroxidase (GPx) levels, especially in younger boys. The exploratory outcome measures failed to show significant effects but there was a trend showing that the younger boys who received treatment were faster at performing the Gowers' maneuver, while the older boys who received treatment were faster at doing the 10-m walk test (10MWT). Therefore, a double-blind, placebo-controlled study for at least two/three years is warranted to verify flavocoxid as a steroid substitute or as add-on therapy to steroids.
Collapse
|
12
|
Cadilla R, Deaton DN, Do Y, Elkins PA, Ennulat D, Guss JH, Holt J, Jeune MR, King AG, Klapwijk JC, Kramer HF, Kramer NJ, Laffan SB, Masuria PI, McDougal AV, Mortenson PN, Musetti C, Peckham GE, Pietrak BL, Poole C, Price DJ, Rendina AR, Sati G, Saxty G, Shearer BG, Shewchuk LM, Sneddon HF, Stewart EL, Stuart JD, Thomas DN, Thomson SA, Ward P, Wilson JW, Xu T, Youngman MA. The exploration of aza-quinolines as hematopoietic prostaglandin D synthase (H-PGDS) inhibitors with low brain exposure. Bioorg Med Chem 2020; 28:115791. [PMID: 33059303 DOI: 10.1016/j.bmc.2020.115791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/15/2022]
Abstract
GlaxoSmithKline and Astex Pharmaceuticals recently disclosed the discovery of the potent H-PGDS inhibitor GSK2894631A 1a (IC50 = 9.9 nM) as part of a fragment-based drug discovery collaboration with Astex Pharmaceuticals. This molecule exhibited good murine pharmacokinetics, allowing it to be utilized to explore H-PGDS pharmacology in vivo. Yet, with prolonged dosing at higher concentrations, 1a induced CNS toxicity. Looking to attenuate brain penetration in this series, aza-quinolines, were prepared with the intent of increasing polar surface area. Nitrogen substitutions at the 6- and 8-positions of the quinoline were discovered to be tolerated by the enzyme. Subsequent structure activity studies in these aza-quinoline scaffolds led to the identification of 1,8-naphthyridine 1y (IC50 = 9.4 nM) as a potent peripherally restricted H-PGDS inhibitor. Compound 1y is efficacious in four in vivo inflammatory models and exhibits no CNS toxicity.
Collapse
Affiliation(s)
- Rodolfo Cadilla
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - David N Deaton
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA.
| | - Young Do
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Patricia A Elkins
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Daniela Ennulat
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jeffrey H Guss
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jason Holt
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Michael R Jeune
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Andrew G King
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Jan C Klapwijk
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - H Fritz Kramer
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Nicholas J Kramer
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Susan B Laffan
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Paresh I Masuria
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Alan V McDougal
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Paul N Mortenson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, UK
| | - Caterina Musetti
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Gregory E Peckham
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Beth L Pietrak
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Chuck Poole
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Daniel J Price
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Alan R Rendina
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Girish Sati
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Gordon Saxty
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, UK
| | - Barry G Shearer
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Lisa M Shewchuk
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Helen F Sneddon
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, UK
| | - Eugene L Stewart
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - J Darren Stuart
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Dean N Thomas
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Stephen A Thomson
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Paris Ward
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Joseph W Wilson
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Tiahshun Xu
- GlaxoSmithKline, 5 Moore Drive, P.O. Box 13398, Research Triangle Park, NC 27709, USA
| | - Mark A Youngman
- GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| |
Collapse
|
13
|
Catapano F, Scaglioni D, Maresh K, Ala P, Domingos J, Selby V, Ricotti V, Phillips L, Servais L, Seferian A, Groot ID, Krom YD, Voit T, Verschuuren JJGM, Niks EH, Straub V, Morgan J, Muntoni F. Novel free-circulating and extracellular vesicle-derived miRNAs dysregulated in Duchenne muscular dystrophy. Epigenomics 2020; 12:1899-1915. [PMID: 33215544 DOI: 10.2217/epi-2020-0052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: To perform cross-sectional and longitudinal miRNA profiling in plasma from Duchenne muscular dystrophy (DMD) subjects and find non-invasive biomarkers in DMD. Subjects/materials & methods: Plasma was collected from 14 age and sex matched controls and 46 DMD subjects. Free-circulating and extracellular vesicle (EV)-derived miRNA expression was measured by RT-qPCR. Results: Free-circulating and EVs derived miR-29c-3p and miR-133a-3p are dysregulated in DMD subjects. Free-circulating and EV-derived miR-29c-3p are reduced in DMD subjects undergoing daily corticosteroid treatment. Free-circulating miR-1-3p and miR-122-5p are longitudinally upregulated in ambulant DMD subjects. Conclusion: We detected novel free-circulating and EV-derived dysregulated miRNAs in plasma from DMD subjects and characterized the longitudinal profile of free-circulating miRNA on plasma from DMD subjects.
Collapse
Affiliation(s)
- Francesco Catapano
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Dominic Scaglioni
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Kate Maresh
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Pierpaolo Ala
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Joana Domingos
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Victoria Selby
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom
| | - Valeria Ricotti
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, United Kingdom
| | - Lauren Phillips
- John Walton Muscular Dystrophy Research Centre, MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Laurent Servais
- Institute I-Motion, Hôpital Armand Trousseau, Paris, France.,Centre de Référence des maladies Neuromusculaires, CHU de Liège, Liège, Belgium
| | | | - Imelda de Groot
- Department of Rehabilitation, Amalia Children's Hospital, Radboud university medical centre, Nijmegen, Netherlands
| | - Yvonne D Krom
- Department of Neurology, Leiden University Medical Center, RC Leiden, Netherlands.,Duchenne Center Netherlands
| | - Thomas Voit
- National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, United Kingdom
| | - J J G M Verschuuren
- Department of Neurology, Leiden University Medical Center, RC Leiden, Netherlands.,Duchenne Center Netherlands
| | - E H Niks
- Department of Neurology, Leiden University Medical Center, RC Leiden, Netherlands.,Duchenne Center Netherlands
| | - Volker Straub
- Northern Genetics Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Institute of Human Genetics, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Jennifer Morgan
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, United Kingdom
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London, WC1N 1EH, United Kingdom.,National Institute for Health Research, Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London WC1N 1EH, United Kingdom
| |
Collapse
|
14
|
Vita GL, Vita G. Is it the right time for an infant screening for Duchenne muscular dystrophy? Neurol Sci 2020; 41:1677-1683. [PMID: 32112218 PMCID: PMC7359158 DOI: 10.1007/s10072-020-04307-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/20/2020] [Indexed: 12/20/2022]
Abstract
Newborn screening (NBS) is an essential, preventive public health programme for early identification of disorders whose early treatment can lead to significant reduction in morbidity and mortality. NBS for Duchenne muscular dystrophy (DMD) has been a controversial matter for many years, because of false positives, the lack of effective drugs and the need of more data about screening efficacy. The still high diagnostic delay of DMD and the current availability of drugs such as steroid, ataluren, eteplirsen, golodirsen and forthcoming new drugs, improving the clinical conditions if early started, make appropriate to begin a concrete discussion between stakeholders to identify best practice for DMD screening. A two-step system CK/DNA screening programme is presented to be performed in male infants aged between 6 months and 42 months involving more than 30,000 male infants. Five to eight DMD subjects are believed to be diagnosed. The pilot project would give the opportunity to test in a small population the feasibility of an infant screening programme, which in the near future could be applicable to an entire country.
Collapse
Affiliation(s)
- Gian Luca Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy
| | - Giuseppe Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy. .,Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| |
Collapse
|
15
|
Hightower RM, Reid AL, Gibbs DE, Wang Y, Widrick JJ, Kunkel LM, Kastenschmidt JM, Villalta SA, van Groen T, Chang H, Gornisiewicz S, Landesman Y, Tamir S, Alexander MS. The SINE Compound KPT-350 Blocks Dystrophic Pathologies in DMD Zebrafish and Mice. Mol Ther 2020; 28:189-201. [PMID: 31628052 PMCID: PMC6952030 DOI: 10.1016/j.ymthe.2019.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/23/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle wasting disease that is caused by the loss of functional dystrophin protein in cardiac and skeletal muscles. DMD patient muscles become weakened, leading to eventual myofiber breakdown and replacement with fibrotic and adipose tissues. Inflammation drives the pathogenic processes through releasing inflammatory cytokines and other factors that promote skeletal muscle degeneration and contributing to the loss of motor function. Selective inhibitors of nuclear export (SINEs) are a class of compounds that function by inhibiting the nuclear export protein exportin 1 (XPO1). The XPO1 protein is an important regulator of key inflammatory and neurological factors that drive inflammation and neurotoxicity in various neurological and neuromuscular diseases. Here, we demonstrate that SINE compound KPT-350 can ameliorate dystrophic-associated pathologies in the muscles of DMD models of zebrafish and mice. Thus, SINE compounds are a promising novel strategy for blocking dystrophic symptoms and could be used in combinatorial treatments for DMD.
Collapse
Affiliation(s)
- Rylie M Hightower
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA; UAB Center for Exercise Medicine (UCEM), Birmingham, AL 35294, USA
| | - Andrea L Reid
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Devin E Gibbs
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA
| | - Yimin Wang
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Jeffrey J Widrick
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics at Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, MA 02115, USA
| | - Jenna M Kastenschmidt
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California-Irvine, Irvine, CA 92697, USA
| | - S Armando Villalta
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California-Irvine, Irvine, CA 92697, USA
| | - Thomas van Groen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hua Chang
- Karyopharm Therapeutics, Newton, MA 02459, USA
| | | | | | | | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA; UAB Center for Exercise Medicine (UCEM), Birmingham, AL 35294, USA; Department of Genetics at the University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center at the University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
16
|
Strandberg K, Ayoglu B, Roos A, Reza M, Niks E, Signorelli M, Fasterius E, Pontén F, Lochmüller H, Domingos J, Ala P, Muntoni F, Aartsma-Rus A, Spitali P, Nilsson P, Szigyarto CAK. Blood-derived biomarkers correlate with clinical progression in Duchenne muscular dystrophy. J Neuromuscul Dis 2020; 7:231-246. [PMID: 32390640 PMCID: PMC7369103 DOI: 10.3233/jnd-190454] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Duchenne Muscular Dystrophy is a severe, incurable disorder caused by mutations in the dystrophin gene. The disease is characterized by decreased muscle function, impaired muscle regeneration and increased inflammation. In a clinical context, muscle deterioration, is evaluated using physical tests and analysis of muscle biopsies, which fail to accurately monitor the disease progression. OBJECTIVES This study aims to confirm and asses the value of blood protein biomarkers as disease progression markers using one of the largest longitudinal collection of samples. METHODS A total of 560 samples, both serum and plasma, collected at three clinical sites are analyzed using a suspension bead array platform to assess 118 proteins targeted by 250 antibodies in microliter amount of samples. RESULTS Nine proteins are confirmed as disease progression biomarkers in both plasma and serum. Abundance of these biomarkers decreases as the disease progresses but follows different trajectories. While carbonic anhydrase 3, microtubule associated protein 4 and collagen type I alpha 1 chain decline rather constantly over time, myosin light chain 3, electron transfer flavoprotein A, troponin T, malate dehydrogenase 2, lactate dehydrogenase B and nestin plateaus in early teens. Electron transfer flavoprotein A, correlates with the outcome of 6-minutes-walking-test whereas malate dehydrogenase 2 together with myosin light chain 3, carbonic anhydrase 3 and nestin correlate with respiratory capacity. CONCLUSIONS Nine biomarkers have been identified that correlate with disease milestones, functional tests and respiratory capacity. Together these biomarkers recapitulate different stages of the disorder that, if validated can improve disease progression monitoring.
Collapse
Affiliation(s)
- Kristin Strandberg
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Burcu Ayoglu
- Department of Protein Science, SciLifeLab, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Andreas Roos
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
- Leibniz-Institut für Analytische Wissenschaften (ISAS), Dortmund, Germany
| | - Mojgan Reza
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Erik Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirko Signorelli
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik Fasterius
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Fredrik Pontén
- Department of Immunology, SciLifeLab, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Hanns Lochmüller
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Neuropediatrics and Muscle Disorders, Medical Center –University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Centro Nacional de Análisis Genómico (CNAGCRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Joana Domingos
- The Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Pierpaolo Ala
- The Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK
- National Institute for Health Research, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, United Kingdom
| | - Annemieke Aartsma-Rus
- MRC Centre for Neuromuscular Diseases, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Nilsson
- Department of Protein Science, SciLifeLab, KTH-Royal Institute of Technology, Stockholm, Sweden
| | - Cristina Al-Khalili Szigyarto
- Department of Protein Science, School of Chemistry, Biotechnology and Health, KTH-Royal Institute of Technology, Stockholm, Sweden
- Department of Protein Science, SciLifeLab, KTH-Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
17
|
Galli F, Bragg L, Meggiolaro L, Rossi M, Caffarini M, Naz N, Santoleri S, Cossu G. Gene and Cell Therapy for Muscular Dystrophies: Are We Getting There? Hum Gene Ther 2019; 29:1098-1105. [PMID: 30132372 PMCID: PMC6211823 DOI: 10.1089/hum.2018.151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In the last few years, significant advances have occurred in the preclinical and clinical work toward gene and cell therapy for muscular dystrophy. At the time of this writing, several trials are ongoing and more are expected to start. It is thus a time of expectation, even though many hurdles remain and it is unclear whether they will be overcome with current strategies or if further improvements will be necessary.
Collapse
Affiliation(s)
- Francesco Galli
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Laricia Bragg
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Linda Meggiolaro
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Maira Rossi
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Miriam Caffarini
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Naila Naz
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Sabrina Santoleri
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| | - Giulio Cossu
- Division of Cell Matrix Biology and Regenerative Medicine, University of Manchester , Manchester, United Kingdom
| |
Collapse
|
18
|
Verhaart IE, Johnson A, Thakrar S, Vroom E, De Angelis F, Muntoni F, Aartsma-Rus AM, Niks EH. Muscle biopsies in clinical trials for Duchenne muscular dystrophy – Patients’ and caregivers’ perspective. Neuromuscul Disord 2019; 29:576-584. [DOI: 10.1016/j.nmd.2019.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/13/2019] [Accepted: 06/05/2019] [Indexed: 12/25/2022]
|
19
|
Ricci F, Vacchetti M, Brusa C, Vercelli L, Davico C, Vitiello B, Mongini T. New pharmacotherapies for genetic neuromuscular disorders: opportunities and challenges. Expert Rev Clin Pharmacol 2019; 12:757-770. [DOI: 10.1080/17512433.2019.1634543] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Federica Ricci
- Division of Child Neurology and Psychiatry, Department of Pediatrics, Regina Margherita Hospital, and Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Martina Vacchetti
- Division of Child Neurology and Psychiatry, Department of Pediatrics, Regina Margherita Hospital, and Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Chiara Brusa
- Dubowitz Neuromuscular Centre, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Liliana Vercelli
- Department of Neuroscience “Rita Levi Montalcini”; Hospital Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Chiara Davico
- Division of Child Neurology and Psychiatry, Department of Pediatrics, Regina Margherita Hospital, and Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Benedetto Vitiello
- Division of Child Neurology and Psychiatry, Department of Pediatrics, Regina Margherita Hospital, and Department of Public Health and Pediatric Sciences, University of Turin, Turin, Italy
| | - Tiziana Mongini
- Department of Neuroscience “Rita Levi Montalcini”; Hospital Città della Salute e della Scienza, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Wang WT, Han C, Sun YM, Chen TQ, Chen YQ. Noncoding RNAs in cancer therapy resistance and targeted drug development. J Hematol Oncol 2019; 12:55. [PMID: 31174564 PMCID: PMC6556047 DOI: 10.1186/s13045-019-0748-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 05/31/2019] [Indexed: 02/08/2023] Open
Abstract
Noncoding RNAs (ncRNAs) represent a large segment of the human transcriptome and have been shown to play important roles in cellular physiology and disease pathogenesis. Increasing evidence on the functional roles of ncRNAs in cancer progression emphasizes the potential of ncRNAs for cancer treatment. Here, we summarize the roles of ncRNAs in disease relapse and resistance to current standard chemotherapy and radiotherapy; the current research progress on ncRNAs for clinical and/or potential translational applications, including the identification of ncRNAs as therapeutic targets; therapeutic approaches for ncRNA targeting; and ncRNA delivery strategies in potential clinical translation. Several ongoing clinical trials of novel RNA-based therapeutics were also emphasized. Finally, we discussed the perspectives and obstacles to different target combinations, delivery strategies, and system designs for ncRNA application. The next approved nucleic acid drug to treat cancer patients may realistically be on the horizon.
Collapse
Affiliation(s)
- Wen-Tao Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Cai Han
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yu-Meng Sun
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Tian-Qi Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China
| | - Yue-Qin Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, Sun Yat-sen University, Guangzhou, 510275, China. .,School of Life Science, Sun Yat-sen University, Guangzhou, 510275, People's Republic of China.
| |
Collapse
|
21
|
|
22
|
Cai A, Kong X. Development of CRISPR-Mediated Systems in the Study of Duchenne Muscular Dystrophy. Hum Gene Ther Methods 2019; 30:71-80. [PMID: 31062609 DOI: 10.1089/hgtb.2018.187] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe type of X-linked recessive degenerative muscle disease caused by mutations in the dystrophin (DMD) gene on the X chromosome. The DMD gene is complex, consisting of 79 exons, and mutations cause changes in the DMD mRNA so that the reading frame is altered, and the muscle-specific isoform of the dystrophin protein is either absent or truncated with variable residual function. The emerging CRISPR-Cas9-mediated genome editing technique is being developed as a potential therapeutic approach to treat DMD because it can permanently replace the mutated dystrophin gene with the normal gene. Prenatal DNA testing can inform whether the female fetus is a carrier of DMD, and the male fetus has inherited a mutation from his mother (50% chance of both). This article summarizes the present status of current and future treatments for DMD.
Collapse
Affiliation(s)
- Aojie Cai
- Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Xiangdong Kong
- Genetic and Prenatal Diagnosis Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
23
|
Sánchez AI, Mariño N, Araujo AF, Espinosa E. Distrofia muscular de Becker con duplicación en el exón 5del gen DMD. REPERTORIO DE MEDICINA Y CIRUGÍA 2019. [DOI: 10.31260/repertmedcir.v28.n2.2019.920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Las distrofinopatías son un grupo de enfermedades ligadas al cromosoma X que abarcan diferentes entidades, siendo las más importantes la distrofia muscular de Duchenne (DMD) y la de Becker (DMB). Están causadas por mutaciones en el gen de la distrofina (gen DMD) localizado en el cromosoma X, locus Xp21.1. En relación con el tipo de mutaciones reportadas en el gen DMD, las delecciones y las mutaciones puntuales son las más comunes, mientras que las duplicaciones corresponden a 10-12%. Aunque las duplicaciones que abarcan el exón 5 ya han sido reportadas en la literatura, a la fecha no existen informes de casos que establezcan una relación genotipo fenotipo clara. Presentamos el caso de un paciente con distrofia muscular de Becker con un fenotipo no tan severo, en quien se encontró una duplicación en el exón 5. Con este caso pretendemos profundizar en la relación genotipo-fenotipo de la DMB, reportando las características clínicas en relación con la duplicación del exón 5 encontrada.
Collapse
|
24
|
Vita G, Vita GL, Musumeci O, Rodolico C, Messina S. Genetic neuromuscular disorders: living the era of a therapeutic revolution. Part 2: diseases of motor neuron and skeletal muscle. Neurol Sci 2019; 40:671-681. [PMID: 30805745 DOI: 10.1007/s10072-019-03764-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/13/2019] [Indexed: 12/22/2022]
Abstract
This is the second part of a two-part document intended to discuss recent therapeutic progresses in genetic neuromuscular disorders. The present review is for diseases of motor neuron and skeletal muscle, some of which reached recently the most innovative therapeutic approaches. Nusinersen, an SMN2 mRNA splicing modifier, was approved as first-ever therapy of spinal muscular atrophy (SMA) by FDA in 2016 and by EMA in 2017. The orally administered small-molecule risdiplam, which increases SMN protein levels similarly but also in peripheral organs, is tested in ongoing phase 2 and 3 trials. After positive results with phase 1 treatment with AAV9-SMN, the first gene therapy for SMA, a phase 3 clinical trial is ongoing. Ataluren is the first approved drug for Duchenne muscular dystrophy (DMD) patients with premature stop codon mutations and its indication has been recently extended since the age of 2 years. Exon skipping technology was and is currently tested in many phase 3 trials, and eteplirsen received a conditional approval by FDA for patients amenable to exon 51 skipping, but not by EMA. Many other compounds with different mechanisms of action are now tested in DMD by phase 2 and 3 trials, including phase 1 gene therapy. Other innovative approaches are under investigation, i.e., gene therapy in X-linked myotubular myopathy and Pompe disease, and antisense oligonucleotides in myotonic dystrophy type 1. Positive evidences are discussed about lamotrigine and ranolazine in non-dystrophic myotonias, chaperons in Pompe disease, and nucleosides in mitochondrial DNA depletion induced by thymidine kinase 2 deficiency.
Collapse
Affiliation(s)
- Giuseppe Vita
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy. .,Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy.
| | - Gian Luca Vita
- Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy
| | - Olimpia Musumeci
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Carmelo Rodolico
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Sonia Messina
- Unit of Neurology and Neuromuscular Diseases, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.,Nemo Sud Clinical Centre for Neuromuscular Disorders, Messina, Italy
| |
Collapse
|
25
|
Li H, Xiao L, Wang L, Lin J, Luo M, Chen M, He R, Zhu Y, Zhang C. HLA Polymorphism Affects Risk of de novo Mutation of dystrophin Gene and Clinical Severity of Duchenne Muscular Dystrophy in a Southern Chinese Population. Front Neurol 2018; 9:970. [PMID: 30498470 PMCID: PMC6249334 DOI: 10.3389/fneur.2018.00970] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 10/29/2018] [Indexed: 01/08/2023] Open
Abstract
Immune-mediated pathology has been thought to be an important factor contributing to Duchenne muscular dystrophy (DMD). Allele frequencies of certain HLA types are known to differ between patients with dystrophinopathies and healthy controls with low-resolution HLA gene typing data in limit reports. Using Polymerase chain reactionsequence based typing (PCR-SBT) to genotype 64 children with DMD in HLA-A, -B,-C, -DRB1, and -DQB1 locus and 503 healthy controls in HLA-A, -B, -DRB1 locus, this study aimed to investigate associations of specific HLA alleles with, and their possible roles in the development and clinical phenotypic severity of DMD. The χ2 test was used to evaluate the distribution of allele frequencies in HLA-A, -B, -DRB1 locus between the patients and healthy controls. A significantly higher frequency of HLA-B*07:05 was found in children with DMD compared to that in controls (OR = 16.2, 95%CI = 2.9–89.3, P < 0.046). More importantly, significantly higher frequencies of HLA-A*29:01 (OR = 77.308, 95%CI = 6.794–879.731, P < 0.0160) and HLA-B*07:05 (OR = 60.240, 95%CI = 9.637–376.535, P < 2.41*10−3) was found in patients with de novo mutations (n = 14) compared to controls while no difference of HLA alleles frequency ware indicated between patients with inherited mutation and control. The result indicates that HLA alleles is associated with pathogenesis of DMD especially DMD with de novo mutation. We use Vignos scale to estimate the lower limb motor function of patients. The impact of HLA alleles on score of Vignos scale of DMD children was estimated by multiple linear regression. Our study indicates that HLA-A*02:01 may have a dampening effect on the clinical phenotypic severity of DMD, evidenced by the presence of HLA-A*02:01 being associated with lower Vignos score. Our study demonstrates that certain HLA alleles are indeed associated with the pathogenesis and clinical phenotypic severity of DMD.
Collapse
Affiliation(s)
- Huan Li
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lulu Xiao
- Department of Tissue Typing Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinfu Lin
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Min Luo
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Menglong Chen
- Department of Neurology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruojie He
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuling Zhu
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Cheng Zhang
- Department of Neurology, National Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
26
|
Messina S, Vita GL. Clinical management of Duchenne muscular dystrophy: the state of the art. Neurol Sci 2018; 39:1837-1845. [PMID: 30218397 DOI: 10.1007/s10072-018-3555-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/04/2018] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a devastating, progressive neuromuscular disorder for which there is no cure. As the dystrophin gene is located on the X chromosome, DMD occurs predominately in males. DMD is caused by a lack of functional dystrophin protein resulting from mutations in the 2.2-Mb DMD gene, whichdisrupts the reading frame. Care considerations for DMD advocate a coordinated, multidisciplinary approach to the management of DMD in order to optimize management of the primary manifestations of DMD as well as any secondary complications that may arise. METHODS This review provides an overview of the multidisciplinary clinical management of DMD with regard to the respiratory, cardiology, orthopedic, and nutritional needs of patients with DMD. Recent advances in novel disease-modifying treatments for DMD are also discussed with specific reference to exon skipping and suppression of premature stop codons as promising genetic therapies. RESULTS The combination of multidisciplinary clinical management alongside novel gene therapiesoffers physicians a powerful armamentarium for the treatment of DMD.
Collapse
Affiliation(s)
- Sonia Messina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy. .,Nemo Sud Clinical Centre, University Hospital "G. Martino", Messina, Italy. .,Unit of Neurology and Neuromuscular Diseases, AOU Policlinico "G. Martino", Building E, 2° Floor, Via Consolare Valeria 1, 98125, Messina, Italy.
| | - Gian Luca Vita
- Nemo Sud Clinical Centre, University Hospital "G. Martino", Messina, Italy
| |
Collapse
|