1
|
Saeed M, Haque A, Shoaib A, Danish Rizvi SM. Exploring novel natural compound-based therapies for Duchenne muscular dystrophy management: insights from network pharmacology, QSAR modeling, molecular dynamics, and free energy calculations. Front Pharmacol 2024; 15:1395014. [PMID: 39415830 PMCID: PMC11481126 DOI: 10.3389/fphar.2024.1395014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 05/31/2024] [Indexed: 10/19/2024] Open
Abstract
Muscular dystrophies encompass a heterogeneous group of rare neuromuscular diseases characterized by progressive muscle degeneration and weakness. Among these, Duchenne muscular dystrophy (DMD) stands out as one of the most severe forms. The present study employs an integrative approach combining network pharmacology, quantitative structure-activity relationship (QSAR) modeling, molecular dynamics (MD) simulations, and free energy calculations to identify potential therapeutic targets and natural compounds for DMD. Upon analyzing the GSE38417 dataset, it was found that individuals with DMD exhibited 290 upregulated differentially expressed genes (DEGs) compared to healthy controls. By utilizing gene ontology (GO) and protein-protein interaction (PPI) network analysis, this study provides insights into the functional roles of the identified DEGs, identifying ten hub genes that play a critical role in the pathology of DMD. These key genes include DMD, TTN, PLEC, DTNA, PKP2, SLC24A, FBXO32, SNTA1, SMAD3, and NOS1. Furthermore, through the use of ligand-based pharmacophore modeling and virtual screening, three natural compounds were identified as potential inhibitors. Among these, compounds 3874518 and 12314417 have demonstrated significant promise as an inhibitor of the SMAD3 protein, a crucial factor in the fibrotic and inflammatory mechanisms associated with DMD. The therapeutic potential of the compounds was further supported by molecular dynamics simulation and Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) analysis. These findings suggest that the compounds are viable candidates for experimental validation against DMD.
Collapse
Affiliation(s)
- Mohd Saeed
- Department of Biology, College of Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Ashanul Haque
- Department of Chemistry, College of Sciences, University of Ha’il, Ha’il, Saudi Arabia
| | - Ambreen Shoaib
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| |
Collapse
|
2
|
Furlong P, Dugar A, White M. Patient engagement in clinical trial design for rare neuromuscular disorders: impact on the DELIVER and ACHIEVE clinical trials. RESEARCH INVOLVEMENT AND ENGAGEMENT 2024; 10:1. [PMID: 38167117 PMCID: PMC10759564 DOI: 10.1186/s40900-023-00535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Engaging individuals living with disease in drug development and regulatory processes leads to more thoughtful and sensitive trial designs, drives more informative and meaningful outcomes from clinical studies, and builds trust between the public, government, and industry stakeholders. This engagement is especially important in the case of rare diseases, where affected individuals and their families face many difficulties getting information, treatment, and support. Dyne Therapeutics is developing therapeutics for people with genetically-driven muscle diseases. During the development of potential treatments for Duchenne muscular dystrophy (DMD) and myotonic dystrophy type 1 (DM1), Dyne sought the opinions of individuals living with these diseases to inform its clinical trial design and to decrease the difficulties that participants and families might experience participating in them. METHODS Dyne engaged individuals and families living with DMD and DM1 as expert partners in its clinical development programs. Dyne convened panels of affected individuals and care partners/parents of individuals living with DMD (n = 8) or DM1 (n = 18). Workshops focused on how affected individuals and their families evaluate and select clinical trials for participation, the importance, quality, and burden associated with individual trial design elements, participation considerations such as site location and the study visit design, patient privacy, the suitability and scope of travel and participant support programs, and the accessibility of content in the informed consent (or assent) forms. Dyne also engaged the DMD Community Advisory Board (CAB) to collect feedback and advice on designing optimal and meaningful clinical trials and measuring relevant outcomes. RESULTS The issues most important to individuals living with DM1 and DMD regarding clinical trials were the ability to participate/access to the trial, perceptions of benefit and risk of trials and potential treatments, the flexibility of participation, clear communication from the sponsor, availability of information from trusted sources, and patient enrollment. In response to the patient advisory workshops and CAB feedback, Dyne refined clinical trial inclusion/exclusion criteria and clinic visit design, developed a travel service program to address the burden of clinical trial travel and enable long-distance and cross-border participation, planned for home visits when feasible, and allowed for adequate rest before clinic visit initiation and between assessments. Additionally, Dyne developed and implemented a transparent and consistent communications plan (including age-appropriate content) for trial participants and community members, and assessed and adjusted procedures to provide maximum participant comfort and lower anxiety, particularly with younger participants. CONCLUSIONS Ongoing communication with the Duchenne CAB and with DMD and DM1 patient advisory committee members allows Dyne to stay current with disease community perspectives and feedback on the needs and preferences of those affected and has provided valuable insights into the participant experience thereby helping Dyne initiate clinical trials that better meet the needs of affected individuals and their families.
Collapse
Affiliation(s)
- Patricia Furlong
- Parent Project Muscular Dystrophy, 1012 14th NW, Suite 500, Washington, DC, 20005, USA.
| | | | | |
Collapse
|
3
|
Wen Y, Yang L, Shen G, Dai S, Wang J, Wang X. A novel splicing mutation identified in a DMD patient: a case report. Front Pediatr 2023; 11:1261318. [PMID: 38054185 PMCID: PMC10694253 DOI: 10.3389/fped.2023.1261318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD, ORPHA:98896) is a lethal X-linked recessive disease that manifests as progressive muscular weakness and wasting. Mutations in the dystrophy gene (DMD) are the main cause of Duchenne muscular dystrophy. Case presentation This study aims to determine novel mutations of DMD and help preimplantation genetic diagnosis (PGD) for family planning. Here present a 4-year-old Chinses boy with DMD, whole-exome sequencing (WES) was performed to identify the molecular basis of the disease. It was confirmed that the boy carried a novel hemizygous mutation of NC_000023.11(NM_004006.3): c.5912_5922 + 19delinsATGTATG in DMD which inherited from his mother. This led to the aberrant splicing of DMD which demonstrated by a minigene splicing assay and further resulted in the impairment of the dystrophy protein. Conclusions Our study discovered a novel splicing mutation of DMD in a DMD patient, which expands the variant spectrum of this gene and provide precise genetic diagnosis of DMD for timely therapy. Meanwhile, this finding will supply valuable information for preimplantation genetic diagnosis.
Collapse
Affiliation(s)
- Yuting Wen
- Department of Obstetrics and Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Luo Yang
- Department of Urology & Pelvic Surgery, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Gan Shen
- Department of Obstetrics and Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Siyu Dai
- Department of Obstetrics and Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Xiang Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Kaslow JA, Sokolow AG, Donnelly T, Buchowski MS, Markham LW, Burnette WB, Soslow JH. Spirometry correlates with physical activity in patients with Duchenne muscular dystrophy. Pediatr Pulmonol 2023; 58:1034-1041. [PMID: 36571207 PMCID: PMC10023371 DOI: 10.1002/ppul.26289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/01/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is associated with progressive muscle weakness and respiratory decline. To date, studies have focused on respiratory decline and loss of ambulation as a metric of loss of skeletal muscle strength. However, new functional measures can assess skeletal muscle disease regardless of ambulatory status. The relationship between these tests and concurrent lung function is currently unexplored. OBJECTIVE To assess the correlation between spirometry measurements and functional muscle assessments such as accelerometry and quantitative muscle testing (QMT). METHODS Enrolled patients with DMD underwent accelerometry and QMT at study clinic visits. Any pulmonary function testing within 6 months of visit was obtained from the electronic medical record. The Spearman correlation coefficient was used to assess the relationship between spirometry and functional muscle testing. RESULTS Forced vital capacity (FVC) and forced expiratory volume in 1 s (FEV1 ) demonstrated the strongest correlation with accelerometry. Both FVC and FEV1 showed a similar relationship to accelerometry when activity was divided into intensity categories, with low intensity and moderate-to-vigorous activity categories showing the strongest correlation. Maximal expiratory pressure (MEP) and FVC showed the most robust correlations with total QMT (p < 0.001 and p < 0.01, respectively). CONCLUSION Lung function, specifically FVC percent predicted and FEV1 %p, shows a good correlation with upper and lower extremity skeletal muscle functional testing such as accelerometry and QMT.
Collapse
Affiliation(s)
- Jacob A Kaslow
- Department of Pediatrics, Division of Pediatric Pulmonary, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew G Sokolow
- Department of Pediatrics, Division of Pediatric Pulmonary, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Thomas Donnelly
- Department of Pediatrics, Thomas P Graham Jr. Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Maciej S Buchowski
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Larry W Markham
- Department of Pediatrics, Division of Cardiology, Indiana University School of Medicine and Riley Hospital for Children at Indiana University Health, Indianapolis, Indiana, USA
| | - William Bryan Burnette
- Division of Pediatric Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jonathan H Soslow
- Department of Pediatrics, Thomas P Graham Jr. Division of Pediatric Cardiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
5
|
He M, Yokota T. Exons 45-55 Skipping Using Antisense Oligonucleotides in Immortalized Human DMD Muscle Cells. Methods Mol Biol 2023; 2640:313-325. [PMID: 36995604 DOI: 10.1007/978-1-0716-3036-5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Antisense oligonucleotides (AOs) have demonstrated high potential as a therapy for treating genetic diseases like Duchene muscular dystrophy (DMD). As a synthetic nucleic acid, AOs can bind to a targeted messenger RNA (mRNA) and regulate splicing. AO-mediated exon skipping transforms out-of-frame mutations as seen in DMD into in-frame transcripts. This exon skipping approach results in the production of a shortened but still functional protein product as seen in the milder counterpart, Becker muscular dystrophy (BMD). Many potential AO drugs have advanced from laboratory experimentation to clinical trials with an increasing interest in this area. An accurate and efficient method for testing AO drug candidates in vitro, before implementation in clinical trials, is crucial to ensure proper assessment of efficacy. The type of cell model used to examine AO drugs in vitro establishes the foundation of the screening process and can significantly impact the results. Previous cell models used to screen for potential AO drug candidates, such as primary muscle cell lines, have limited proliferative and differentiation capacity, and express insufficient amounts of dystrophin. Recently developed immortalized DMD muscle cell lines effectively addressed this challenge allowing for the accurate measurement of exon-skipping efficacy and dystrophin protein production. This chapter presents a procedure used to assess DMD exons 45-55 skipping efficiency and dystrophin protein production in immortalized DMD patient-derived muscle cells. Exons 45-55 skipping in the DMD gene is potentially applicable to 47% of patients. In addition, naturally occurring exons 45-55 in-frame deletion mutation is associated with an asymptomatic or remarkably mild phenotype as compared to shorter in-frame deletions within this region. As such, exons 45-55 skipping is a promising therapeutic approach to treat a wider group of DMD patients. The method presented here allows for improved examination of potential AO drugs before implementation in clinical trials for DMD.
Collapse
Affiliation(s)
- Merry He
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
6
|
Sulfur amino acid supplementation displays therapeutic potential in a C. elegans model of Duchenne muscular dystrophy. Commun Biol 2022; 5:1255. [PMID: 36385509 PMCID: PMC9668843 DOI: 10.1038/s42003-022-04212-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 11/01/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the dystrophin gene cause Duchenne muscular dystrophy (DMD), a common muscle disease that manifests with muscle weakness, wasting, and degeneration. An emerging theme in DMD pathophysiology is an intramuscular deficit in the gasotransmitter hydrogen sulfide (H2S). Here we show that the C. elegans DMD model displays reduced levels of H2S and expression of genes required for sulfur metabolism. These reductions can be offset by increasing bioavailability of sulfur containing amino acids (L-methionine, L-homocysteine, L-cysteine, L-glutathione, and L-taurine), augmenting healthspan primarily via improved calcium regulation, mitochondrial structure and delayed muscle cell death. Additionally, we show distinct differences in preservation mechanisms between sulfur amino acid vs H2S administration, despite similarities in required health-preserving pathways. Our results suggest that the H2S deficit in DMD is likely caused by altered sulfur metabolism and that modulation of this pathway may improve DMD muscle health via multiple evolutionarily conserved mechanisms. A C. elegans model of Duchenne muscular dystrophy reveals a potential role for disrupted sulfur metabolism in the disease and thus the therapeutic potential of sulfur amino acid supplementation.
Collapse
|
7
|
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
|
8
|
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
|
9
|
Effects of Low-Intensity and Long-Term Aerobic Exercise on the Psoas Muscle of mdx Mice: An Experimental Model of Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:ijms23094483. [PMID: 35562874 PMCID: PMC9105402 DOI: 10.3390/ijms23094483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/27/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscle disease characterized by the absence of the protein dystrophin, which causes a loss of sarcolemma integrity, determining recurrent muscle injuries, decrease in muscle function, and progressive degeneration. Currently, there is a need for therapeutic treatments to improve the quality of life of DMD patients. Here, we investigated the effects of a low-intensity aerobic training (37 sessions) on satellite cells, peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α protein (PGC-1α), and different types of fibers of the psoas muscle from mdx mice (DMD experimental model). Wildtype and mdx mice were randomly divided into sedentary and trained groups (n = 24). Trained animals were subjected to 37 sessions of low-intensity running on a motorized treadmill. Subsequently, the psoas muscle was excised and analyzed by immunofluorescence for dystrophin, satellite cells, myosin heavy chain (MHC), and PGC-1α content. The minimal Feret’s diameters of the fibers were measured, and light microscopy was applied to observe general morphological features of the muscles. The training (37 sessions) improved morphological features in muscles from mdx mice and caused an increase in the number of quiescent/activated satellite cells. It also increased the content of PGC-1α in the mdx group. We concluded that low-intensity aerobic exercise (37 sessions) was able to reverse deleterious changes determined by DMD.
Collapse
|
10
|
Kotov AA, Bazylev SS, Adashev VE, Shatskikh AS, Olenina LV. Drosophila as a Model System for Studying of the Evolution and Functional Specialization of the Y Chromosome. Int J Mol Sci 2022; 23:4184. [PMID: 35457001 PMCID: PMC9031259 DOI: 10.3390/ijms23084184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023] Open
Abstract
The Y chromosome is one of the sex chromosomes found in males of animals of different taxa, including insects and mammals. Among all chromosomes, the Y chromosome is characterized by a unique chromatin landscape undergoing dynamic evolutionary change. Being entirely heterochromatic, the Y chromosome as a rule preserves few functional genes, but is enriched in tandem repeats and transposons. Due to difficulties in the assembly of the highly repetitive Y chromosome sequence, deep analyses of Y chromosome evolution, structure, and functions are limited to a few species, one of them being Drosophila melanogaster. Despite Y chromosomes exhibiting high structural divergence between even closely related species, Y-linked genes have evolved convergently and are mainly associated with spermatogenesis-related activities. This indicates that male-specific selection is a dominant force shaping evolution of Y chromosomes across species. This review presents our analysis of current knowledge concerning Y chromosome functions, focusing on recent findings in Drosophila. Here we dissect the experimental and bioinformatics data about the Y chromosome accumulated to date in Drosophila species, providing comparative analysis with mammals, and discussing the relevance of our analysis to a wide range of eukaryotic organisms, including humans.
Collapse
Affiliation(s)
| | | | | | | | - Ludmila V. Olenina
- Institute of Molecular Genetics of National Research Center «Kurchatov Institute», 123182 Moscow, Russia; (A.A.K.); (S.S.B.); (V.E.A.); (A.S.S.)
| |
Collapse
|
11
|
Lingineni K, Aggarwal V, Morales JF, Conrado DJ, Corey D, Vong C, Burton J, Larkindale J, Romero K, Schmidt S, Kim S. Development of a model-based clinical trial simulation platform to optimize the design of clinical trials for Duchenne muscular dystrophy. CPT Pharmacometrics Syst Pharmacol 2022; 11:318-332. [PMID: 34877803 PMCID: PMC8923721 DOI: 10.1002/psp4.12753] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/25/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022] Open
Abstract
Early clinical trials of therapies to treat Duchenne muscular dystrophy (DMD), a fatal genetic X‐linked pediatric disease, have been designed based on the limited understanding of natural disease progression and variability in clinical measures over different stages of the continuum of the disease. The objective was to inform the design of DMD clinical trials by developing a disease progression model‐based clinical trial simulation (CTS) platform based on measures commonly used in DMD trials. Data were integrated from past studies through the Duchenne Regulatory Science Consortium founded by the Critical Path Institute (15 clinical trials and studies, 1505 subjects, 27,252 observations). Using a nonlinear mixed‐effects modeling approach, longitudinal dynamics of five measures were modeled (NorthStar Ambulatory Assessment, forced vital capacity, and the velocities of the following three timed functional tests: time to stand from supine, time to climb 4 stairs, and 10 meter walk‐run time). The models were validated on external data sets and captured longitudinal changes in the five measures well, including both early disease when function improves as a result of growth and development and the decline in function in later stages. The models can be used in the CTS platform to perform trial simulations to optimize the selection of inclusion/exclusion criteria, selection of measures, and other trial parameters. The data sets and models have been reviewed by the US Food and Drug Administration and the European Medicines Agency; have been accepted into the Fit‐for‐Purpose and Qualification for Novel Methodologies pathways, respectively; and will be submitted for potential endorsement by both agencies.
Collapse
Affiliation(s)
- Karthik Lingineni
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | | - Diane Corey
- Critical Path Institute, Tucson, Arizona, USA
| | - Camille Vong
- Global Product Development, Pfizer Inc, Cambridge, Massachusetts, USA
| | | | | | | | - Stephan Schmidt
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | | |
Collapse
|
12
|
Roshmi RR, Yokota T. Pharmacological Profile of Viltolarsen for the Treatment of Duchenne Muscular Dystrophy: A Japanese Experience. Clin Pharmacol 2021; 13:235-242. [PMID: 34938127 PMCID: PMC8688746 DOI: 10.2147/cpaa.s288842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/12/2021] [Indexed: 01/11/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal, X-linked recessive disorder characterized by progressive muscle loss and cardiorespiratory complications. Mutations in the DMD gene that eliminate the production of dystrophin protein are the underlying causes of DMD. Viltolarsen is a drug of phosphorodiamidate morpholino oligomer (PMO) chemistry, designed to skip exon 53 of the DMD gene. It aims to produce truncated but partially functional dystrophin in DMD patients and restore muscle function. Based on a preclinical study showing the ability of antisense PMOs targeting the DMD gene to improve muscle function in a large animal model, viltolarsen was developed by Nippon Shinyaku and the National Center of Neurology and Psychiatry in Japan. Following clinical trials conducted in Japan, Canada, and the United States showing significant improvements in muscle function, viltolarsen was approved for medical use in Japan in March 2020 and the United States in August 2020, respectively. Viltolarsen is a mutation-specific drug and will work for 8% of the persons with DMD who carry mutations amenable to exon 53 skipping. This review summarizes the pharmacological profile of viltolarsen, important clinical trials, and challenges, focusing on the contribution of Japanese patients and researchers in its development.
Collapse
Affiliation(s)
- Rohini Roy Roshmi
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
13
|
Gaina G, Vossen RHAM, Manole E, Plesca DA, Ionica E. Combining Protein Expression and Molecular Data Improves Mutation Characterization of Dystrophinopathies. Front Neurol 2021; 12:718396. [PMID: 34950096 PMCID: PMC8689184 DOI: 10.3389/fneur.2021.718396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Duchenne and Becker muscular dystrophy are X-linked recessive inherited disorders characterized by progressive weakness due to skeletal muscle degeneration. Different mutations in the DMD gene, which encodes for dystrophin protein, are responsible for these disorders. The aim of our study was to investigate the relationship between type, size, and location of the mutation that occurs in the DMD gene and their effect on dystrophin protein expression in a cohort of 40 male dystrophinopathy patients and nine females, possible carriers. We evaluated the expression of dystrophin by immunofluorescence and immunoblotting. The mutational spectrum of the DMD gene was established by MLPA for large copy number variants, followed by HRM analysis for point mutations and sequencing of samples with an abnormal melting profile. MLPA revealed 30 deletions (75%) and three duplications (7.5%). HRM analysis accounted for seven-point mutations (17.5%). We also report four novel small mutations (c. 8507G>T, c.3021delG, c.9563_9563+1insAGCATGTTTATGATACAGCA, c.7661-60T>A) in DMD gene. Our work shows that the DNA translational open reading frame and the location of the mutation both influence the expression of dystrophin and disease severity phenotype. The proposed algorithm used in this study demonstrates its accuracy for the characterization of dystrophinopathy patients.
Collapse
Affiliation(s)
- Gisela Gaina
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, Bucharest, Romania
- *Correspondence: Gisela Gaina ;
| | - Rolf H. A. M. Vossen
- Center for Human and Clinical Genetics, Leiden Genome Technology Center, Leiden, Netherlands
| | - Emilia Manole
- Laboratory of Cell Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, Bucharest, Romania
- Colentina Clinical Hospital, Bucharest, Romania
| | - Doina Anca Plesca
- Department of Pediatrics, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Clinical Pediatrics, Victor Gomoiu Children Clinical Hospital, Bucharest, Romania
| | - Elena Ionica
- Department of Biochemistry and Molecular Biology, University of Bucharest, Bucharest, Romania
| |
Collapse
|
14
|
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
|
15
|
Triana-Fonseca P, Parada-Márquez JF, Silva-Aldana CT, Zambrano-Arenas D, Arias-Gomez LL, Morales-Fonseca N, Medina-Méndez E, Restrepo CM, Silgado-Guzmán DF, Fonseca-Mendoza DJ. Genetic Profile of the Dystrophin Gene Reveals New Mutations in Colombian Patients Affected with Muscular Dystrophinopathy. APPLICATION OF CLINICAL GENETICS 2021; 14:399-408. [PMID: 34629887 PMCID: PMC8493106 DOI: 10.2147/tacg.s317721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/31/2021] [Indexed: 11/23/2022]
Abstract
Background Duchenne and Becker muscular dystrophies (DMD/BMD) are the most common human dystrophinopathies with recessive X-linked inheritance. Dystrophin gene deletions and duplications are the most common mutations, followed by point mutations. The aim of this study is to characterize the mutational profile of the dystrophin gene in Colombian patients with DMD/BMD. Material and Methods Mutational profiling was determined in 69 affected patients using Sanger sequencing, next-generation sequencing (NGS) and/or multiplex ligation dependent-probes amplification (MLPA). Genetic variants were classified according to molecular consequence and new variants were determined through database and literature analysis. Results Mutational profile in affected patients revealed that large deletions/duplications analyzed by MLPA accounted for 72.5% of all genetic variations. By using Sanger sequencing or NGS, we identified point mutations in 15.9% and small deletions in 11.6% of the patients. New mutations were found, most of them were point mutations or small deletions (10.1%). Conclusion Our results described the genetic profile of the dystrophin gene in Colombian patients with DMD and contribute to efforts to identify molecular variants in Latin American populations. For our population, 18.8% of cases could be treated with FDA or MDA approved molecular therapies based on specific mutations. These data contribute to the establishment of appropriate genetic counseling and potential treatment.
Collapse
Affiliation(s)
| | | | - Claudia T Silva-Aldana
- Department of Molecular Diagnosis, Genética Molecular de Colombia SAS, Bogotá, DC, Colombia
| | | | | | | | - Esteban Medina-Méndez
- Department of Molecular Diagnosis, Genética Molecular de Colombia SAS, Bogotá, DC, Colombia
| | - Carlos M Restrepo
- Center for Research in Genetics and Genomics - CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, DC, Colombia
| | | | - Dora Janeth Fonseca-Mendoza
- Center for Research in Genetics and Genomics - CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad Del Rosario, Bogotá, DC, Colombia
| |
Collapse
|
16
|
Nocetti PM, Alberti A, Freiberger V, Ventura L, Grigollo LR, Andreau CS, Júnior RJN, Martins DF, Comim CM. Swimming Improves Memory and Antioxidant Defense in an Animal Model of Duchenne Muscular Dystrophy. Mol Neurobiol 2021; 58:5067-5077. [PMID: 34245442 DOI: 10.1007/s12035-021-02482-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/01/2021] [Indexed: 01/15/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease associated with progressive skeletal muscle degeneration. In humans, DMD has an early onset, causes developmental delays, and is a devastating disease that drastically diminishes the quality of life of young individuals affected. The objective of this study was to evaluate the effects of a swimming protocol on memory and oxidative stress in an animal model of DMD. Male mdx and wild-type mice aged ≥ 28 days were used in this study. The animals were trained for a stepped swimming protocol for four consecutive weeks. The swimming protocol significantly reduced the levels of lipid peroxidation and protein carbonylation in the gastrocnemius, hippocampus, and striatum in the exercised animals. It also prevented lipid peroxidation in the diaphragm. Moreover, it increased the free thiol levels in the gastrocnemius, the diaphragm, and all central nervous system structures. The results showed that the protocol that applied swimming as a low-intensity aerobic exercise for 4 weeks prevented aversive memory and habituation in mdx mice.
Collapse
Affiliation(s)
- Priscila Mantovani Nocetti
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| | - Adriano Alberti
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil.
| | - Viviane Freiberger
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| | - Letícia Ventura
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| | - Leoberto Ricardo Grigollo
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| | - Cristina Salar Andreau
- CEU Cardenal Herrera University, Carrer Lluís Vives, 1, 46115, Valencia, Alfara del Patriarca, Spain
| | - Rudy José Nodari Júnior
- Department of Physical Education, University of West Santa Catarina, Área das Ciências Biológicas E da Saúde, Rua José Firmo Bernardi, 1591, Bairro Flor da Serra, Joaçaba, Santa Catarina, 89600-000, Brazil
| | - Daniel Fernandes Martins
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| | - Clarissa M Comim
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Cidade Universitária Pedra Branca, 88137-272, Palhoça, SC, Brazil
| |
Collapse
|
17
|
Ellwood RA, Piasecki M, Szewczyk NJ. Caenorhabditis elegans as a Model System for Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22094891. [PMID: 34063069 PMCID: PMC8125261 DOI: 10.3390/ijms22094891] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
The nematode worm Caenorhabditis elegans has been used extensively to enhance our understanding of the human neuromuscular disorder Duchenne Muscular Dystrophy (DMD). With new arising clinically relevant models, technologies and treatments, there is a need to reconcile the literature and collate the key findings associated with this model.
Collapse
Affiliation(s)
- Rebecca A. Ellwood
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Mathew Piasecki
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence:
| |
Collapse
|
18
|
Niu X, Menhart N. Structural Perturbations of Exon-Skipping Edits within the Dystrophin D20:24 Region. Biochemistry 2021; 60:765-779. [PMID: 33656846 DOI: 10.1021/acs.biochem.0c00827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Exon skipping is a disease-modifying therapy in which oligonucleotide analogues mask specific exons, eliminating them from the mature mRNA, and also the cognate protein. That is one possible therapeutic aim, but it can also be used to restore the reading frame for diseases caused by frameshift mutations, which is the case for Duchenne muscular dystrophy (DMD). DMD most commonly arises as a result of large exonic deletions that create a frameshift and abolish protein expression. Loss of dystrophin protein leads to the pathology of the disease, which is severe, causing death generally in the second or third decade of life. Here, the primary aim of exon skipping is restoration of protein expression by reading frame correction. However, the therapeutically expressed protein is missing both the region of the underlying genetic defect and the therapeutically skipped exon. How removing some region from the middle of a protein affects its structure and function is unclear. Many different underlying deletions are known, and exon skipping can be applied in many ways, in some cases in different ways to the same defect. These vary in how severely perturbative they are, with possible clinical consequences. In this study, we examine a systematic, comprehensive panel of exon edits in a region of dystrophin and identify for the first time exon edits that are minimally perturbed and appear to keep the structural stability similar to that of wild-type protein. We also identify factors that appear to be correlated with how perturbative an edit is.
Collapse
Affiliation(s)
- Xin Niu
- Department of Biology, Illinois Institute of Technology, 3101 South Dearborn Street, Chicago, Illinois 60616, United States
| | - Nick Menhart
- Department of Biology, Illinois Institute of Technology, 3101 South Dearborn Street, Chicago, Illinois 60616, United States
| |
Collapse
|
19
|
Botzenhart UU, Keil C, Tsagkari E, Zeidler-Rentzsch I, Gredes T, Gedrange T. Influence of botulinum toxin A on craniofacial morphology after injection into the right masseter muscle of dystrophin deficient (mdx-) mice. Ann Anat 2021; 236:151715. [PMID: 33675949 DOI: 10.1016/j.aanat.2021.151715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND Severe craniofacial and dental abnormalities, typical for patients with progressive Duchenne muscular dystrophy (DMD), are an exellcent demonstration of Melvin L. Moss "functional matrix theory", highlighting the influence of muscle tissue on craniofacial growth and morphology. However, the currently best approved animal model for investigation of this interplay is the mdx-mouse, which offers only a limited time window for research, due to the ability of muscle regeneration, in contrast to the human course of the disease. The aim of this study was to evaluate craniofacial morphology after BTX-A induced muscle paralysis in C57Bl- and mdx-mice, to prove the suitability of BTX-A intervention to inhibit muscle regeneration in mdx-mice and thus, mimicking the human course of the DMD disease. METHODS Paralysis of the right masseter muscle was induced in 100 days old C57Bl- and mdx-mice by a single specific intramuscular BTX-A injection. Mice skulls were obtained at 21 days and 42 days after BTX-A injection and 3D radiological evaluation was performed in order to measure various craniofacial dimensions in the sagittal, transversal and vertical plane. Statstical analysis were performed using SigmaStat®Version 3.5. In case of normal distribution, unpaired t-test and otherwise the Mann-Whitney-U test was applied. A statistical significance was given in case of p ≤ 0.05. RESULTS In contrast to C57Bl-mice, in mdx-mice, three weeks after BTX-A treatment a significant decrease of skull dimensions was noted in most of the measurements followed by a significant increase at the second investigation period. CONCLUSIONS BTX-A can induce changes in craniofacial morphology and presumably partially inhibit muscle regeneration in mdx-mice, but cannot completely intensify craniofacial effects elicited by dystrophy. Further research is necessary in order to fully understand muscle-bone interplay after BTX-A injection into dystrophic muscles.
Collapse
Affiliation(s)
| | - Christiane Keil
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany; Department of Orthodontics, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Eirini Tsagkari
- Department of Orthodontics, Faculty of Dentistry School of Health Sciences, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Ines Zeidler-Rentzsch
- Department of Otorhinolaryngology, Head and Neck Surgery, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Tomasz Gredes
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany; Department of Orthodontics, Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| | - Tomasz Gedrange
- Medical Faculty Carl Gustav Carus Campus, TU Dresden, 01307, Dresden, Germany
| |
Collapse
|
20
|
Ellwood RA, Hewitt JE, Torregrossa R, Philp AM, Hardee JP, Hughes S, van de Klashorst D, Gharahdaghi N, Anupom T, Slade L, Deane CS, Cooke M, Etheridge T, Piasecki M, Antebi A, Lynch GS, Philp A, Vanapalli SA, Whiteman M, Szewczyk NJ. Mitochondrial hydrogen sulfide supplementation improves health in the C. elegans Duchenne muscular dystrophy model. Proc Natl Acad Sci U S A 2021; 118:e2018342118. [PMID: 33627403 PMCID: PMC7936346 DOI: 10.1073/pnas.2018342118] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder characterized by progressive muscle degeneration and weakness due to mutations in the dystrophin gene. The symptoms of DMD share similarities with those of accelerated aging. Recently, hydrogen sulfide (H2S) supplementation has been suggested to modulate the effects of age-related decline in muscle function, and metabolic H2S deficiencies have been implicated in affecting muscle mass in conditions such as phenylketonuria. We therefore evaluated the use of sodium GYY4137 (NaGYY), a H2S-releasing molecule, as a possible approach for DMD treatment. Using the dys-1(eg33) Caenorhabditis elegans DMD model, we found that NaGYY treatment (100 µM) improved movement, strength, gait, and muscle mitochondrial structure, similar to the gold-standard therapeutic treatment, prednisone (370 µM). The health improvements of either treatment required the action of the kinase JNK-1, the transcription factor SKN-1, and the NAD-dependent deacetylase SIR-2.1. The transcription factor DAF-16 was required for the health benefits of NaGYY treatment, but not prednisone treatment. AP39 (100 pM), a mitochondria-targeted H2S compound, also improved movement and strength in the dys-1(eg33) model, further implying that these improvements are mitochondria-based. Additionally, we found a decline in total sulfide and H2S-producing enzymes in dystrophin/utrophin knockout mice. Overall, our results suggest that H2S deficit may contribute to DMD pathology, and rectifying/overcoming the deficit with H2S delivery compounds has potential as a therapeutic approach to DMD treatment.
Collapse
MESH Headings
- Animals
- Caenorhabditis elegans/genetics
- Caenorhabditis elegans/metabolism
- Caenorhabditis elegans Proteins/genetics
- Caenorhabditis elegans Proteins/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dystrophin/deficiency
- Dystrophin/genetics
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Gene Expression Regulation
- Humans
- Hydrogen Sulfide/metabolism
- Hydrogen Sulfide/pharmacology
- Locomotion/drug effects
- Locomotion/genetics
- Male
- Mice
- Mice, Inbred mdx
- Mitochondria, Muscle/drug effects
- Mitochondria, Muscle/metabolism
- Mitochondria, Muscle/pathology
- Mitogen-Activated Protein Kinases/genetics
- Mitogen-Activated Protein Kinases/metabolism
- Morpholines/metabolism
- Morpholines/pharmacology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/drug therapy
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Organophosphorus Compounds/metabolism
- Organophosphorus Compounds/pharmacology
- Organothiophosphorus Compounds/metabolism
- Organothiophosphorus Compounds/pharmacology
- Prednisone/pharmacology
- Sirtuins/genetics
- Sirtuins/metabolism
- Thiones/metabolism
- Thiones/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Utrophin/deficiency
- Utrophin/genetics
Collapse
Affiliation(s)
- Rebecca A Ellwood
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby DE22 3DT, United Kingdom
| | - Jennifer E Hewitt
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Roberta Torregrossa
- University of Exeter Medical School, University of Exeter, EX1 2LU Exeter, United Kingdom
| | - Ashleigh M Philp
- Mitochondrial Metabolism and Ageing, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent's Clinical School, University of New South Wales (UNSW) Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Justin P Hardee
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Samantha Hughes
- HAN BioCentre, HAN University of Applied Sciences, Nijmegen 6525EM, The Netherlands
| | | | - Nima Gharahdaghi
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby DE22 3DT, United Kingdom
| | - Taslim Anupom
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX 79409
| | - Luke Slade
- University of Exeter Medical School, University of Exeter, EX1 2LU Exeter, United Kingdom
- Sport and Health Sciences, University of Exeter, EX1 2LU Exeter, United Kingdom
| | - Colleen S Deane
- Sport and Health Sciences, University of Exeter, EX1 2LU Exeter, United Kingdom
- Living System Institute, University of Exeter, EX4 4QD Exeter, United Kingdom
| | - Michael Cooke
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby DE22 3DT, United Kingdom
- Sport and Health Sciences, University of Exeter, EX1 2LU Exeter, United Kingdom
| | - Timothy Etheridge
- Sport and Health Sciences, University of Exeter, EX1 2LU Exeter, United Kingdom
| | - Mathew Piasecki
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby DE22 3DT, United Kingdom
| | - Adam Antebi
- Molecular Genetics of Ageing, Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Andrew Philp
- Mitochondrial Metabolism and Ageing, Garvan Institute of Medical Research, Sydney, NSW 2010, Australia
- St. Vincent's Clinical School, University of New South Wales (UNSW) Medicine, University of New South Wales Sydney, Sydney, NSW 2052, Australia
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409
| | - Matthew Whiteman
- University of Exeter Medical School, University of Exeter, EX1 2LU Exeter, United Kingdom;
| | - Nathaniel J Szewczyk
- Medical Research Council (MRC) Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom;
- Musculoskeletal Conditions, National Institute for Health Research Nottingham Biomedical Research Centre, Derby DE22 3DT, United Kingdom
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH 45701
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701
| |
Collapse
|
21
|
Sun C, Shen L, Zhang Z, Xie X. Therapeutic Strategies for Duchenne Muscular Dystrophy: An Update. Genes (Basel) 2020; 11:genes11080837. [PMID: 32717791 PMCID: PMC7463903 DOI: 10.3390/genes11080837] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022] Open
Abstract
Neuromuscular disorders encompass a heterogeneous group of conditions that impair the function of muscles, motor neurons, peripheral nerves, and neuromuscular junctions. Being the most common and most severe type of muscular dystrophy, Duchenne muscular dystrophy (DMD), is caused by mutations in the X-linked dystrophin gene. Loss of dystrophin protein leads to recurrent myofiber damage, chronic inflammation, progressive fibrosis, and dysfunction of muscle stem cells. Over the last few years, there has been considerable development of diagnosis and therapeutics for DMD, but current treatments do not cure the disease. Here, we review the current status of DMD pathogenesis and therapy, focusing on mutational spectrum, diagnosis tools, clinical trials, and therapeutic approaches including dystrophin restoration, gene therapy, and myogenic cell transplantation. Furthermore, we present the clinical potential of advanced strategies combining gene editing, cell-based therapy with tissue engineering for the treatment of muscular dystrophy.
Collapse
Affiliation(s)
- Chengmei Sun
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining 314400, China; (C.S.); (L.S.); (Z.Z.)
- Department of Medical Oncology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Luoan Shen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining 314400, China; (C.S.); (L.S.); (Z.Z.)
| | - Zheng Zhang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining 314400, China; (C.S.); (L.S.); (Z.Z.)
| | - Xin Xie
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining 314400, China; (C.S.); (L.S.); (Z.Z.)
- Department of Medical Oncology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
- Correspondence: ; Tel.: +86-0571-87572326
| |
Collapse
|
22
|
Hoepers A, Alberti A, Freiberger V, Ventura L, Grigollo LR, Andreu CS, da Silva BB, Martins DF, Junior RJN, Streck EL, Comim CM. Effect of Aerobic Physical Exercise in an Animal Model of Duchenne Muscular Dystrophy. J Mol Neurosci 2020; 70:1552-1564. [PMID: 32507928 DOI: 10.1007/s12031-020-01565-0] [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: 10/14/2019] [Accepted: 04/22/2020] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a condition caused by an amendment to the X chromosome, inherited as a recessive trait, and affects 1:3500 live births, especially males. Low-intensity exercise is known to decrease certain parameters associated with muscular degeneration in animal models of progressive muscular dystrophies. In the present study, 28-day-old male mdx and wild-type (wild) mice were used. The animals were subjected to a low-intensity physical exercise protocol for 8 weeks. It was found that this protocol was able to reduce oxidative stress in muscle tissue and in most of the CNS structures analyzed, with a significant increase in antioxidant activity in all analyzed structures. It is thus possible to infer that this exercise protocol was able to reduce oxidative stress and improve the energy metabolism in brain tissue and in the gastrocnemius muscle of animals with DMD.
Collapse
Affiliation(s)
- Andreza Hoepers
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | - Adriano Alberti
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil.
| | - Viviane Freiberger
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | - Letícia Ventura
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | - Leoberto Ricardo Grigollo
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | | | - Bruna Becker da Silva
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | - Daniel Fernandes Martins
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| | | | - Emilio L Streck
- Laboratory of Experimental Physiopathology, Postgraduate Program in Health Sciences, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - Clarissa M Comim
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences University of South Santa Catarina, Palhoça, SC, Brazil
| |
Collapse
|
23
|
Heterogenetic parabiosis between healthy and dystrophic mice improve the histopathology in muscular dystrophy. Sci Rep 2020; 10:7075. [PMID: 32341395 PMCID: PMC7184587 DOI: 10.1038/s41598-020-64042-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 04/09/2020] [Indexed: 11/10/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle disease, characterized by mutations in the X-linked dystrophin, that has several therapeutic options but no curative treatment. Transplantation of muscle progenitor cells for treatment of DMD has been widely investigated; however, its application is hindered by limited cell survival due to the harmful dystrophic microenvironment. An alternative approach to utilize progenitor cells and circulatory factors and to improve the dystrophic muscle pathology and microenvironment is through parabiotic pairing, where mice are surgically sutured to create a joint circulatory system. Parabiotic mice were generated by surgically joining wild type (WT) mice expressing green fluorescent protein (GFP) with mdx mice. These mice developed a common circulation (approximately 50% green cells in the blood of mdx mice) 2-weeks after parabiotic pairing. We observed significantly improved dystrophic muscle pathology, including decreased inflammation, necrotic fibers and fibrosis in heterogenetic parabionts. Importantly, the GFP + cells isolated from the mdx mice (paired with GFP mice) underwent myogenic differentiation in vitro and expressed markers of mesenchymal stem cells and macrophages, which may potentially be involved in the improvement of dystrophic muscle pathology. These observations suggest that changing the dystrophic microenvironment can be a new approach to treat DMD.
Collapse
|
24
|
Morelli KH, Hatton CL, Harper SQ, Burgess RW. Gene therapies for axonal neuropathies: Available strategies, successes to date, and what to target next. Brain Res 2020; 1732:146683. [PMID: 32001243 DOI: 10.1016/j.brainres.2020.146683] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/20/2022]
Abstract
Nearly one-hundred loci in the human genome have been associated with different forms of Charcot-Marie-Tooth disease (CMT) and related inherited neuropathies. Despite this wealth of gene targets, treatment options are still extremely limited, and clear "druggable" pathways are not obvious for many of these mutations. However, recent advances in gene therapies are beginning to circumvent this challenge. Each type of CMT is a monogenic disorder, and the cellular targets are usually well-defined and typically include peripheral neurons or Schwann cells. In addition, the genetic mechanism is often also clear, with loss-of-function mutations requiring restoration of gene expression, and gain-of-function or dominant-negative mutations requiring silencing of the mutant allele. These factors combine to make CMT a good target for developing genetic therapies. Here we will review the state of relatively established gene therapy approaches, including viral vector-mediated gene replacement and antisense oligonucleotides for exon skipping, altering splicing, and gene knockdown. We will also describe earlier stage approaches for allele-specific knockdown and CRIPSR/Cas9 gene editing. We will next describe how these various approaches have been deployed in clinical and preclinical studies. Finally, we will evaluate various forms of CMT as candidates for gene therapy based on the current understanding of their genetics, cellular/tissue targets, validated animal models, and availability of patient populations and natural history data.
Collapse
Affiliation(s)
- Kathryn H Morelli
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | | | - Scott Q Harper
- 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
| | - Robert W Burgess
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; The Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
25
|
Transcriptomic Analysis Reveals Involvement of the Macrophage Migration Inhibitory Factor Gene Network in Duchenne Muscular Dystrophy. Genes (Basel) 2019; 10:genes10110939. [PMID: 31752120 PMCID: PMC6896047 DOI: 10.3390/genes10110939] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 01/04/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive hereditary muscular disease with X-linked recessive inheritance, that leads patients to premature death. The loss of dystrophin determines membrane instability, causing cell damage and inflammatory response. Macrophage migration inhibitory factor (MIF) is a cytokine that exerts pleiotropic properties and is implicated in the pathogenesis of a variety of diseases. Recently, converging data from independent studies have pointed to a possible role of MIF in dystrophic muscle disorders, including DMD. In the present study, we have investigated the modulation of MIF and MIF-related genes in degenerative muscle disorders, by making use of publicly available whole-genome expression datasets. We show here a significant enrichment of MIF and related genes in muscle samples from DMD patients, as well as from patients suffering from Becker’s disease and limb-girdle muscular dystrophy type 2B. On the other hand, transcriptomic analysis of in vitro differentiated myotubes from healthy controls and DMD patients revealed no significant alteration in the expression levels of MIF-related genes. Finally, by analyzing DMD samples as a time series, we show that the modulation of the genes belonging to the MIF network is an early event in the DMD muscle and does not change with the increasing age of the patients, Overall, our analysis suggests that MIF may play a role in vivo during muscle degeneration, likely promoting inflammation and local microenvironment reaction.
Collapse
|
26
|
Boehler JF, Ricotti V, Gonzalez JP, Soustek-Kramer M, Such L, Brown KJ, Schneider JS, Morris CA. Membrane recruitment of nNOSµ in microdystrophin gene transfer to enhance durability. Neuromuscul Disord 2019; 29:735-741. [PMID: 31521486 DOI: 10.1016/j.nmd.2019.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/25/2022]
Abstract
Several gene transfer clinical trials are currently ongoing with the common aim of delivering a shortened version of dystrophin, termed a microdystrophin, for the treatment of Duchenne muscular dystrophy (DMD). However, one of the main differences between these trials is the microdystrophin protein produced following treatment. Each gene transfer product is based on different selections of dystrophin domain combinations to assemble microdystrophin transgenes that maintain functional dystrophin domains and fit within the packaging limits of an adeno-associated virus (AAV) vector. While domains involved in mechanical function, such as the actin-binding domain and β-dystroglycan binding domain, have been identified for many years and included in microdystrophin constructs, more recently the neuronal nitric oxide synthase (nNOS) domain has also been identified due to its role in enhancing nNOS membrane localization. As nNOS membrane localization has been established as an important requirement for prevention of functional ischemia in skeletal muscle, inclusion of the nNOS domain into a microdystrophin construct represents an important consideration. The aim of this mini review is to highlight what is currently known about the nNOS domain of dystrophin and to describe potential implications of this domain in a microdystrophin gene transfer clinical trial.
Collapse
Affiliation(s)
- Jessica F Boehler
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Valeria Ricotti
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - J Patrick Gonzalez
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | | | - Lauren Such
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Kristy J Brown
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Joel S Schneider
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States
| | - Carl A Morris
- Solid Biosciences, 141 Portland Street, Cambridge, MA 02139, United States.
| |
Collapse
|
27
|
Egorova TV, Zotova ED, Reshetov DA, Polikarpova AV, Vassilieva SG, Vlodavets DV, Gavrilov AA, Ulianov SV, Buchman VL, Deykin AV. CRISPR/Cas9-generated mouse model of Duchenne muscular dystrophy recapitulating a newly identified large 430 kb deletion in the human DMD gene. Dis Model Mech 2019; 12:dmm037655. [PMID: 31028078 PMCID: PMC6505476 DOI: 10.1242/dmm.037655] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/20/2019] [Indexed: 01/10/2023] Open
Abstract
Exon skipping is a promising strategy for Duchenne muscular dystrophy (DMD) disease-modifying therapy. To make this approach safe, ensuring that excluding one or more exons will restore the reading frame and that the resulting protein will retain critical functions of the full-length dystrophin protein is necessary. However, in vivo testing of the consequences of skipping exons that encode the N-terminal actin-binding domain (ABD) has been confounded by the absence of a relevant animal model. We created a mouse model of the disease recapitulating a novel human mutation, a large de novo deletion of exons 8-34 of the DMD gene, found in a Russian DMD patient. This mutation was achieved by deleting exons 8-34 of the X-linked mouse D md gene using CRISPR/Cas9 genome editing, which led to a reading frame shift and the absence of functional dystrophin production. Male mice carrying this deletion display several important signs of muscular dystrophy, including a gradual age-dependent decrease in muscle strength, increased creatine kinase, muscle fibrosis and central nucleation. The degrees of these changes are comparable to those observed in mdx mice, a standard laboratory model of DMD. This new model of DMD will be useful for validating therapies based on skipping exons that encode the N-terminal ABD and for improving our understanding of the role of the N-terminal domain and central rod domain in the biological function of dystrophin. Simultaneous skipping of exons 6 and 7 should restore the gene reading frame and lead to the production of a protein that might retain functionality despite the partial deletion of the ABD.
Collapse
Affiliation(s)
- Tatiana V Egorova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Marlin Biotech LLC, Moscow, 143026, Russia
| | | | | | - Anna V Polikarpova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Marlin Biotech LLC, Moscow, 143026, Russia
| | - Svetlana G Vassilieva
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Marlin Biotech LLC, Moscow, 143026, Russia
| | - Dmitry V Vlodavets
- Veltischev Scientific Research Clinical Paediatric Institute, Moscow, 125412, Russia
| | - Alexey A Gavrilov
- Group of Genome Spatial Organization, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Sergey V Ulianov
- Laboratory of Structural and Functional Organization of Chromosomes, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, 119991, Russia
| | | | - Alexei V Deykin
- Core Facilities, Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| |
Collapse
|
28
|
Ma KM, Thomas ES, Wereszczynski J, Menhart N. Empirical and Computational Comparison of Alternative Therapeutic Exon Skip Repairs for Duchenne Muscular Dystrophy. Biochemistry 2019; 58:2061-2076. [PMID: 30896926 DOI: 10.1021/acs.biochem.9b00062] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a common and devastating genetic disease primarily caused by exon deletions that create a genetic frameshift in dystrophin. Exon skipping therapy seeks to correct this by masking an exon during the mRNA maturation process, restoring dystrophin expression, but creating an edited protein missing both the original defect and the therapeutically skipped region. Crucially, it is possible to correct many defects in alternative ways, by skipping an exon either before or after the patient's defect. This results in alternatively edited, hybrid proteins that might have different properties and therapeutic consequences. We examined three such dystrophin exon-skipped edits, Δe45-53, Δe46-54, and Δe47-55, comprising two pairs of alternative repairs of Δe46-53 and Δe47-54 DMD defects. We found that in both cases, Δe46-54 was the more stable repair as determined by a variety of thermodynamic and biochemical measurements. We also examined the origin of these differences with molecular dynamics simulations, which showed that these stability differences were the result of different types of structural perturbations. For example, in one edit there was partial unfolding at the edit site that caused domain-localized perturbations while in another there was unfolding at the protein domain junctions distal to the edit site that increased molecular flexibility. These results demonstrate that alternative exon skip repairs of the same underlying defect can have very different consequences at the level of protein structure and stability and furthermore that these can arise by different mechanisms, either locally or by more subtle long-range perturbations.
Collapse
|
29
|
Hewitt JE, Pollard AK, Lesanpezeshki L, Deane CS, Gaffney CJ, Etheridge T, Szewczyk NJ, Vanapalli SA. Muscle strength deficiency and mitochondrial dysfunction in a muscular dystrophy model of Caenorhabditis elegans and its functional response to drugs. Dis Model Mech 2018; 11:dmm036137. [PMID: 30396907 PMCID: PMC6307913 DOI: 10.1242/dmm.036137] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/30/2018] [Indexed: 12/31/2022] Open
Abstract
Muscle strength is a key clinical parameter used to monitor the progression of human muscular dystrophies, including Duchenne and Becker muscular dystrophies. Although Caenorhabditis elegans is an established genetic model for studying the mechanisms and treatments of muscular dystrophies, analogous strength-based measurements in this disease model are lacking. Here, we describe the first demonstration of the direct measurement of muscular strength in dystrophin-deficient C. elegans mutants using a micropillar-based force measurement system called NemaFlex. We show that dys-1(eg33) mutants, but not dys-1(cx18) mutants, are significantly weaker than their wild-type counterparts in early adulthood, cannot thrash in liquid at wild-type rates, display mitochondrial network fragmentation in the body wall muscles, and have an abnormally high baseline mitochondrial respiration. Furthermore, treatment with prednisone, the standard treatment for muscular dystrophy in humans, and melatonin both improve muscular strength, thrashing rate and mitochondrial network integrity in dys-1(eg33), and prednisone treatment also returns baseline respiration to normal levels. Thus, our results demonstrate that the dys-1(eg33) strain is more clinically relevant than dys-1(cx18) for muscular dystrophy studies in C. elegans This finding, in combination with the novel NemaFlex platform, can be used as an efficient workflow for identifying candidate compounds that can improve strength in the C. elegans muscular dystrophy model. Our study also lays the foundation for further probing of the mechanism of muscle function loss in dystrophin-deficient C. elegans, leading to knowledge translatable to human muscular dystrophy.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Jennifer E Hewitt
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Amelia K Pollard
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham & National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Leila Lesanpezeshki
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Colleen S Deane
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
| | - Christopher J Gaffney
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
- Lancaster Medical School, Furness College, Lancaster University, Lancaster LA1 4YG, UK
| | - Timothy Etheridge
- Sport and Health Sciences, University of Exeter, St Luke's Campus, Exeter EX1 2LU, UK
| | - Nathaniel J Szewczyk
- MRC/ARUK Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham & National Institute for Health Research Nottingham Biomedical Research Centre, Derby, UK
| | - Siva A Vanapalli
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
30
|
Lee MMY, McMurray JJV, Lorenzo-Almorós A, Kristensen SL, Sattar N, Jhund PS, Petrie MC. Diabetic cardiomyopathy. Heart 2018; 105:337-345. [PMID: 30337334 DOI: 10.1136/heartjnl-2016-310342] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Matthew Meng Yang Lee
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - John J V McMurray
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ana Lorenzo-Almorós
- Renal, Vascular and Diabetes Laboratory, Instituto de Investigaciónes Sanitarias-Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
| | - Søren Lund Kristensen
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Pardeep S Jhund
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
31
|
Denti F, Paludan-Müller C, Olesen SP, Haunsø S, Svendsen JH, Olesen MS, Bentzen BH, Schmitt N. Functional consequences of genetic variation in sodium channel modifiers in early onset lone atrial fibrillation. Per Med 2018; 15:93-102. [DOI: 10.2217/pme-2017-0076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim: We investigated the effect of variants in genes encoding sodium channel modifiers SNTA1 and GPD1L found in early onset atrial fibrillation (AF) patients. Patients & methods: Genetic screening in patients with early onset lone AF revealed three variants in GPD1L and SNTA1 in three AF patients. Functional analysis was performed by patch-clamp electrophysiology. Results: Co-expression of GPD1L or its p.A326E variant with NaV1.5 did not alter INa density or current kinetics. SNTA1 shifted the peak-current by -5 mV. The SNTA1-p.A257G variant significantly increased INa. SNTA1-p.P74L did not produce functional changes. Conclusion: Although genetic variation of sodium channel modifiers may contribute to development of AF at a molecular level, it is unlikely a monogenic cause of the disease.
Collapse
Affiliation(s)
- Federico Denti
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Paludan-Müller
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Søren-Peter Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stig Haunsø
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health & Medical Sciences, University of Copenhagen, Denmark
| | - Jesper Hastrup Svendsen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health & Medical Sciences, University of Copenhagen, Denmark
| | - Morten Salling Olesen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Laboratory for Molecular Cardiology, Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Bo Hjorth Bentzen
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nicole Schmitt
- Danish National Research Foundation Centre for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health & Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
32
|
Duchêne B, Iyombe-Engembe JP, Rousseau J, Tremblay JP, Ouellet DL. From gRNA Identification to the Restoration of Dystrophin Expression: A Dystrophin Gene Correction Strategy for Duchenne Muscular Dystrophy Mutations Using the CRISPR-Induced Deletion Method. Methods Mol Biol 2018; 1687:267-283. [PMID: 29067670 DOI: 10.1007/978-1-4939-7374-3_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
The discovery of the CRISPR-Cas9 system raises hope for the treatment of many genetic disorders. We describe here an approach based on the use of a pair of single guide RNAs to form a hybrid exon that does not only restore the dystrophin gene reading frame but also results in the production of a dystrophin protein with an adequate structure of the central rod-domain, with a correct spectrin-like repeat. The therapeutic approach described here involved DMD patient cells having a deletion of exons 51-53 of the DMD gene.
Collapse
Affiliation(s)
- Benjamin Duchêne
- Centre de Recherche du CHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC, Canada, G1V 4G2
| | - Jean-Paul Iyombe-Engembe
- Centre de Recherche du CHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC, Canada, G1V 4G2
| | - Joël Rousseau
- Centre de Recherche du CHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC, Canada, G1V 4G2
| | - Jacques P Tremblay
- Centre de Recherche du CHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC, Canada, G1V 4G2
| | - Dominique L Ouellet
- Centre de Recherche du CHU de Québec-Université Laval, 2705 Boul. Laurier, Québec, QC, Canada, G1V 4G2.
| |
Collapse
|
33
|
Anderson J, Seol H, Gordish-Dressman H, Hathout Y, Spurney CF. Interleukin 1 Receptor-Like 1 Protein (ST2) is a Potential Biomarker for Cardiomyopathy in Duchenne Muscular Dystrophy. Pediatr Cardiol 2017; 38:1606-1612. [PMID: 28821969 PMCID: PMC6317901 DOI: 10.1007/s00246-017-1703-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/28/2017] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a rare, fatal X-linked disorder characterized by the lack of dystrophin, a key sarcolemma muscle protein. Cardiac failure is a significant cause of death in DMD subjects. The purpose of our research was to identify potential cardiac serum biomarkers associated with DMD cardiomyopathy. This is an observational, case-controlled study using subjects from the CINRG DMD natural history study with cardiomyopathy (ejection fraction (EF) <55%; shortening fraction (SF) <28%), subjects without cardiomyopathy (EF ≥ 55%; SF ≥ 28%) compared to normal healthy volunteer subjects. The DMD with cardiomyopathy group had significantly lower average EF and SF (EF = 45 ± 10/SF = 25 ± 2%) than the DMD without cardiomyopathy group (EF = 58 ± 5% and SF = 32 ± 3%; p < 0.01). Among a selected set of potential biomarkers for cardiomyopathy (MMP9, BNP, GAL3, CRP, LEP, TNC, TLR4 and ST2) we validated ST2 as significantly elevated in the serum of DMD cardiomyopathy group (35,798 ± 4884 pg/mL) compared to normal controls (9940 ± 2680 pg/mL; p < 0.01; n = 6). Matrix metallopeptidase 9 (MMP9) levels were found significantly increased in both DMD groups compared to controls (p < 0.01). No significant differences were seen in BNP, GAL3, CRP, LEP, TNC or TLR4 levels. Increased ST2 levels were found in serum of DMD subjects compared to healthy volunteers and further elevated in DMD subjects with cardiomyopathy. Future studies correlating cardiomyopathy with ST2 levels may allow for improved non-invasive monitoring of cardiac disease in DMD subjects.
Collapse
Affiliation(s)
- Julia Anderson
- Research Center for Genetic Medicine, Children’s Research Institute, Children’s National Health System, Washington, DC, United States of America
| | - Haeri Seol
- Research Center for Genetic Medicine, Children’s Research Institute, Children’s National Health System, Washington, DC, United States of America
| | - Heather Gordish-Dressman
- Research Center for Genetic Medicine, Children’s Research Institute, Children’s National Health System, Washington, DC, United States of America
| | - Yetrib Hathout
- Research Center for Genetic Medicine, Children’s Research Institute, Children’s National Health System, Washington, DC, United States of America
| | - Christopher F. Spurney
- Research Center for Genetic Medicine, Children’s Research Institute, Children’s National Health System, Washington, DC, United States of America,Children’s National Heart Institute, Division of Cardiology, Children’s National Health System, Washington, DC, United States of America,Corresponding author: Phone: 202-476-2020,
| | | |
Collapse
|
34
|
Wilson K, Faelan C, Patterson-Kane JC, Rudmann DG, Moore SA, Frank D, Charleston J, Tinsley J, Young GD, Milici AJ. Duchenne and Becker Muscular Dystrophies: A Review of Animal Models, Clinical End Points, and Biomarker Quantification. Toxicol Pathol 2017; 45:961-976. [PMID: 28974147 DOI: 10.1177/0192623317734823] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are neuromuscular disorders that primarily affect boys due to an X-linked mutation in the DMD gene, resulting in reduced to near absence of dystrophin or expression of truncated forms of dystrophin. Some newer therapeutic interventions aim to increase sarcolemmal dystrophin expression, and accurate dystrophin quantification is critical for demonstrating pharmacodynamic relationships in preclinical studies and clinical trials. Current challenges with measuring dystrophin include the variation in protein expression within individual muscle fibers and across whole muscle samples, the presence of preexisting dystrophin-positive revertant fibers, and trace amounts of residual dystrophin. Immunofluorescence quantification of dystrophin can overcome many of these challenges, but manual quantification of protein expression may be complicated by variations in the collection of images, reproducible scoring of fluorescent intensity, and bias introduced by manual scoring of typically only a few high-power fields. This review highlights the pathology of DMD and BMD, discusses animal models of DMD and BMD, and describes dystrophin biomarker quantitation in DMD and BMD, with several image analysis approaches, including a new automated method that evaluates protein expression of individual muscle fibers.
Collapse
Affiliation(s)
- Kristin Wilson
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | - Crystal Faelan
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | | | | | - Steven A Moore
- 2 Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Diane Frank
- 3 Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Jay Charleston
- 3 Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Jon Tinsley
- 4 Summit Therapeutics, Abingdon, United Kingdom
| | - G David Young
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | | |
Collapse
|
35
|
Kyrychenko V, Kyrychenko S, Tiburcy M, Shelton JM, Long C, Schneider JW, Zimmermann WH, Bassel-Duby R, Olson EN. Functional correction of dystrophin actin binding domain mutations by genome editing. JCI Insight 2017; 2:95918. [PMID: 28931764 DOI: 10.1172/jci.insight.95918] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/15/2017] [Indexed: 12/26/2022] Open
Abstract
Dystrophin maintains the integrity of striated muscles by linking the actin cytoskeleton with the cell membrane. Duchenne muscular dystrophy (DMD) is caused by mutations in the dystrophin gene (DMD) that result in progressive, debilitating muscle weakness, cardiomyopathy, and a shortened lifespan. Mutations of dystrophin that disrupt the amino-terminal actin-binding domain 1 (ABD-1), encoded by exons 2-8, represent the second-most common cause of DMD. In the present study, we compared three different strategies for CRISPR/Cas9 genome editing to correct mutations in the ABD-1 region of the DMD gene by deleting exons 3-9, 6-9, or 7-11 in human induced pluripotent stem cells (iPSCs) and by assessing the function of iPSC-derived cardiomyocytes. All three exon deletion strategies enabled the expression of truncated dystrophin protein and restoration of cardiomyocyte contractility and calcium transients to varying degrees. We show that deletion of exons 3-9 by genomic editing provides an especially effective means of correcting disease-causing ABD-1 mutations. These findings represent an important step toward eventual correction of common DMD mutations and provide a means of rapidly assessing the expression and function of internally truncated forms of dystrophin-lacking portions of ABD-1.
Collapse
Affiliation(s)
- Viktoriia Kyrychenko
- Department of Molecular Biology.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sergii Kyrychenko
- Department of Molecular Biology.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - John M Shelton
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Chengzu Long
- Department of Molecular Biology.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jay W Schneider
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - Rhonda Bassel-Duby
- Department of Molecular Biology.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Eric N Olson
- Department of Molecular Biology.,Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, and.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
36
|
Influence of Botulinumtoxin A on the Expression of Adult MyHC Isoforms in the Masticatory Muscles in Dystrophin-Deficient Mice (Mdx-Mice). BIOMED RESEARCH INTERNATIONAL 2016; 2016:7063093. [PMID: 27689088 PMCID: PMC5023834 DOI: 10.1155/2016/7063093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/27/2016] [Accepted: 07/10/2016] [Indexed: 11/17/2022]
Abstract
The most widespread animal model to investigate Duchenne muscular dystrophy is the mdx-mouse. In contrast to humans, phases of muscle degeneration are replaced by regeneration processes; hence there is only a restricted time slot for research. The aim of the study was to investigate if an intramuscular injection of BTX-A is able to break down muscle regeneration and has direct implications on the gene expression of myosin heavy chains in the corresponding treated and untreated muscles. Therefore, paralysis of the right masseter muscle was induced in adult healthy and dystrophic mice by a specific intramuscular injection of BTX-A. After 21 days the mRNA expression and protein content of MyHC isoforms of the right and left masseter, temporal, and the tongue muscle were determined using quantitative RT-PCR and Western blot technique. MyHC-IIa and MyHC-I-mRNA expression significantly increased in the paralyzed masseter muscle of control-mice, whereas MyHC-IIb and MyHC-IIx/d-mRNA were decreased. In dystrophic muscles no effect of BTX-A could be detected at the level of MyHC. This study suggests that BTX-A injection is a suitable method to simulate DMD-pathogenesis in healthy mice but further investigations are necessary to fully analyse the BTX-A effect and to generate sustained muscular atrophy in mdx-mice.
Collapse
|