1
|
McDowall S, Aung-Htut M, Wilton S, Li D. Antisense oligonucleotides and their applications in rare neurological diseases. Front Neurosci 2024; 18:1414658. [PMID: 39376536 PMCID: PMC11456401 DOI: 10.3389/fnins.2024.1414658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/20/2024] [Indexed: 10/09/2024] Open
Abstract
Rare diseases affect almost 500 million people globally, predominantly impacting children and often leading to significantly impaired quality of life and high treatment costs. While significant contributions have been made to develop effective treatments for those with rare diseases, more rapid drug discovery strategies are needed. Therapeutic antisense oligonucleotides can modulate target gene expression with high specificity through various mechanisms determined by base sequences and chemical modifications; and have shown efficacy in clinical trials for a few rare neurological conditions. Therefore, this review will focus on the applications of antisense oligonucleotides, in particular splice-switching antisense oligomers as promising therapeutics for rare neurological diseases, with key examples of Duchenne muscular dystrophy and spinal muscular atrophy. Challenges and future perspectives in developing antisense therapeutics for rare conditions including target discovery, antisense chemical modifications, animal models for therapeutic validations, and clinical trial designs will also be briefly discussed.
Collapse
Affiliation(s)
- Simon McDowall
- School of Human Sciences, The University of Western Australia, Crawley, WA, Australia
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
| | - May Aung-Htut
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Steve Wilton
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| | - Dunhui Li
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Murdoch, WA, Australia
| |
Collapse
|
2
|
Nakashima M, Suga N, Yoshikawa S, Matsuda S. Caveolin and NOS in the Development of Muscular Dystrophy. Int J Mol Sci 2024; 25:8771. [PMID: 39201459 PMCID: PMC11354531 DOI: 10.3390/ijms25168771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 09/02/2024] Open
Abstract
Caveolin is a structural protein within caveolae that may be involved in transmembrane molecular transport and/or various intercellular interactions within cells. Specific mutations of caveolin-3 in muscle fibers are well known to cause limb-girdle muscular dystrophy. Altered expression of caveolin-3 has also been detected in Duchenne muscular dystrophy, which may be a part of the pathological process leading to muscle weakness. Interestingly, it has been shown that the renovation of nitric oxide synthase (NOS) in sarcolemma with muscular dystrophy could improve muscle health, suggesting that NOS may be involved in the pathology of muscular dystrophy. Here, we summarize the notable function of caveolin and/or NOS in skeletal muscle fibers and discuss their involvement in the pathology as well as possible tactics for the innovative treatment of muscular dystrophies.
Collapse
Affiliation(s)
| | | | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
3
|
Ikelaar NA, Barnard AM, Eng SWM, Hosseini Vajargah S, Ha KCH, Kan HE, Vandenborne K, Niks EH, Walter GA, Spitali P. Large scale serum proteomics identifies proteins associated with performance decline and clinical milestones in Duchenne muscular dystrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.05.24311516. [PMID: 39148831 PMCID: PMC11326316 DOI: 10.1101/2024.08.05.24311516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Serum biomarkers are promising minimally invasive outcome measures in clinical studies in Duchenne muscular dystrophy (DMD). However, biomarkers strongly associated with clinical progression and predicting performance decline are lacking. In this study we aimed to identify serum biomarkers associated with clinical performance and able to predict clinical milestones in DMD. Towards this aim we present a retrospective multi-center cohort study including serum samples and clinical data collected in research participants with DMD as part of a natural history study at the University of Florida (UF) and real-world observations at Leiden University Medical Center (LUMC) between 2009-2022. The 7K SomaScan® assay was used to analyse protein levels in in individual serum samples. Serum biomarkers predicted age at loss of ambulation (LoA), age at loss of overhead reach (OHR) and age at loss of hand to mouth function (HTM). Secondary outcomes were the association of biomarkers with age, corticosteroid (CS) usage, and clinical performance based on the North Star Ambulatory Assessment (NSAA), 10 meter run velocity (10mrv), 6 minute walk (6MWT) and Performance of the Upper Limb (PUL2.0). A total of 716 serum samples were collected in 79 participants at UF and 74 at LUMC (mean[SD] age; 10.9[3.2] vs 8.4[3.4]). 244 serum proteins showed an association with CS usage in both cohorts independent of CS type and regimen, including MMP3 and IGLL1. 318 probes (corresponding to 294 proteins) showed significant associations with NSAA, 10mrv, 6MWT and/or PUL2.0 across both cohorts. The expression of 38 probes corresponding to 36 proteins such as RGMA, EHMT2, ART3, ANTXR2 and DLK1 was associated with risk of both lower and upper limb clinical milestones in both the LUMC and UF cohort. In conclusion, multiple biomarkers were associated with CS use, motor function and upper lower and upper limb disease milestones in DMD. These biomarkers were validated across two independent cohorts, increasing their likelihood of translation for use within the broader DMD population.
Collapse
Affiliation(s)
- N A Ikelaar
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, the Netherlands
- Duchenne Center Netherlands
| | - A M Barnard
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - S W M Eng
- BioSymetrics, Inc., Huntington, NY, USA
| | | | - K C H Ha
- BioSymetrics, Inc., Huntington, NY, USA
| | - H E Kan
- Duchenne Center Netherlands
- C.J. Gorter MRI Center, Department of Radiology, Leiden University Medical Center, Leiden, Zuid-Holland, the Netherlands
| | - K Vandenborne
- Department of Physical Therapy, University of Florida, Gainesville, FL, USA
| | - E H Niks
- Department of Neurology, Leiden University Medical Center, Leiden, Zuid-Holland, the Netherlands
- Duchenne Center Netherlands
| | - G A Walter
- Department of Physiology and Aging, University of Florida, Gainesville, FL, USA
| | - P Spitali
- Human Genetics Department, Leiden University Medical Center, Leiden, Zuid-Holland, the Netherlands
| |
Collapse
|
4
|
Konieczny P. Systemic Treatment of Body-Wide Duchenne Muscular Dystrophy Symptoms. Clin Pharmacol Ther 2024. [PMID: 38965715 DOI: 10.1002/cpt.3363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/09/2024] [Indexed: 07/06/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked disease that leads to premature death due to the loss of dystrophin. Current strategies predominantly focus on the therapeutic treatment of affected skeletal muscle tissue. However, certain results point to the fact that with successful treatment of skeletal muscle, DMD-exposed latent phenotypes in tissues, such as cardiac and smooth muscle, might lead to adverse effects and even death. Likewise, it is now clear that the absence of dystrophin affects the function of the nervous system, and that this phenotype is more pronounced when shorter dystrophins are absent, in addition to the full-length dystrophin that is present predominantly in the muscle. Here, I focus on the systemic aspects of DMD, highlighting the ubiquitous expression of the dystrophin gene in human tissues. Furthermore, I describe therapeutic strategies that have been tested in the clinic and point to unresolved questions regarding the function of distinct dystrophin isoforms, and the possibility of current therapeutic strategies to tackle phenotypes that relate to their absence.
Collapse
Affiliation(s)
- Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, Poznań, Poland
| |
Collapse
|
5
|
Ó Murchú SC, O'Halloran KD. BREATHE DMD: boosting respiratory efficacy after therapeutic hypoxic episodes in Duchenne muscular dystrophy. J Physiol 2024; 602:3255-3272. [PMID: 38837229 DOI: 10.1113/jp280280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/12/2024] [Indexed: 06/07/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal genetic neuromuscular disorder, characterised by progressive decline in skeletal muscle function due to the secondary consequences of dystrophin deficiency. Weakness extends to the respiratory musculature, and cardiorespiratory failure is the leading cause of death in men with DMD. Intermittent hypoxia has emerged as a potential therapy to counteract ventilatory insufficiency by eliciting long-term facilitation of breathing. Mechanisms of sensory and motor facilitation of breathing have been well delineated in animal models. Various paradigms of intermittent hypoxia have been designed and implemented in human trials culminating in clinical trials in people with spinal cord injury and amyotrophic lateral sclerosis. Application of therapeutic intermittent hypoxia to DMD is considered together with discussion of the potential barriers to progression owing to the complexity of this devastating disease. Notwithstanding the considerable challenges and potential pitfalls of intermittent hypoxia-based therapies for DMD, we suggest it is incumbent on the research community to explore the potential benefits in pre-clinical models. Intermittent hypoxia paradigms should be implemented to explore the proclivity to express respiratory plasticity with the longer-term aim of preserving and potentiating ventilation in pre-clinical models and people with DMD.
Collapse
Affiliation(s)
- Seán C Ó Murchú
- Department of Physiology, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
6
|
Morais P, Zhang R, Yu YT. Therapeutic Nonsense Suppression Modalities: From Small Molecules to Nucleic Acid-Based Approaches. Biomedicines 2024; 12:1284. [PMID: 38927491 PMCID: PMC11201248 DOI: 10.3390/biomedicines12061284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/29/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Nonsense mutations are genetic mutations that create premature termination codons (PTCs), leading to truncated, defective proteins in diseases such as cystic fibrosis, neurofibromatosis type 1, Dravet syndrome, Hurler syndrome, Beta thalassemia, inherited bone marrow failure syndromes, Duchenne muscular dystrophy, and even cancer. These mutations can also trigger a cellular surveillance mechanism known as nonsense-mediated mRNA decay (NMD) that degrades the PTC-containing mRNA. The activation of NMD can attenuate the consequences of truncated, defective, and potentially toxic proteins in the cell. Since approximately 20% of all single-point mutations are disease-causing nonsense mutations, it is not surprising that this field has received significant attention, resulting in a remarkable advancement in recent years. In fact, since our last review on this topic, new examples of nonsense suppression approaches have been reported, namely new ways of promoting the translational readthrough of PTCs or inhibiting the NMD pathway. With this review, we update the state-of-the-art technologies in nonsense suppression, focusing on novel modalities with therapeutic potential, such as small molecules (readthrough agents, NMD inhibitors, and molecular glue degraders); antisense oligonucleotides; tRNA suppressors; ADAR-mediated RNA editing; targeted pseudouridylation; and gene/base editing. While these various modalities have significantly advanced in their development stage since our last review, each has advantages (e.g., ease of delivery and specificity) and disadvantages (manufacturing complexity and off-target effect potential), which we discuss here.
Collapse
Affiliation(s)
- Pedro Morais
- Drug Metabolism and Pharmacokinetics, Research and Development, Bayer Pharmaceuticals, 42113 Wuppertal, Germany
| | - Rui Zhang
- Center for RNA Biology, Department of Biochemistry and Biophysics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA;
| | - Yi-Tao Yu
- Center for RNA Biology, Department of Biochemistry and Biophysics, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA;
| |
Collapse
|
7
|
Dhoke NR, Kim H, Azzag K, Crist SB, Kiley J, Perlingeiro RCR. A Novel CRISPR-Cas9 Strategy to Target DYSTROPHIN Mutations Downstream of Exon 44 in Patient-Specific DMD iPSCs. Cells 2024; 13:972. [PMID: 38891104 PMCID: PMC11171783 DOI: 10.3390/cells13110972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Mutations in the DMD gene cause fatal Duchenne Muscular Dystrophy (DMD). An attractive therapeutic approach is autologous cell transplantation utilizing myogenic progenitors derived from induced pluripotent stem cells (iPSCs). Given that a significant number of DMD mutations occur between exons 45 and 55, we developed a gene knock-in approach to correct any mutations downstream of exon 44. We applied this approach to two DMD patient-specific iPSC lines carrying mutations in exons 45 and 51 and confirmed mini-DYSTROPHIN (mini-DYS) protein expression in corrected myotubes by western blot and immunofluorescence staining. Transplantation of gene-edited DMD iPSC-derived myogenic progenitors into NSG/mdx4Cv mice produced donor-derived myofibers, as shown by the dual expression of human DYSTROPHIN and LAMIN A/C. These findings further provide proof-of-concept for the use of programmable nucleases for the development of autologous iPSC-based therapy for muscular dystrophies.
Collapse
Affiliation(s)
- Neha R. Dhoke
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Hyunkee Kim
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Karim Azzag
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Sarah B. Crist
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - James Kiley
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
| | - Rita C. R. Perlingeiro
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA; (N.R.D.); (H.K.); (K.A.); (S.B.C.); (J.K.)
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Kiełbowski K, Bakinowska E, Procyk G, Ziętara M, Pawlik A. The Role of MicroRNA in the Pathogenesis of Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:6108. [PMID: 38892293 PMCID: PMC11172814 DOI: 10.3390/ijms25116108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/29/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked progressive disorder associated with muscle wasting and degeneration. The disease is caused by mutations in the gene that encodes dystrophin, a protein that links the cytoskeleton with cell membrane proteins. The current treatment methods aim to relieve the symptoms of the disease or partially rescue muscle functionality. However, they are insufficient to suppress disease progression. In recent years, studies have uncovered an important role for non-coding RNAs (ncRNAs) in regulating the progression of numerous diseases. ncRNAs, such as micro-RNAs (miRNAs), bind to their target messenger RNAs (mRNAs) to suppress translation. Understanding the mechanisms involving dysregulated miRNAs can improve diagnosis and suggest novel treatment methods for patients with DMD. This review presents the available evidence on the role of altered expression of miRNAs in the pathogenesis of DMD. We discuss the involvement of these molecules in the processes associated with muscle physiology and DMD-associated cardiomyopathy.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Grzegorz Procyk
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Marta Ziętara
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.Z.)
| |
Collapse
|
9
|
Verhaeg M, Adamzek K, van de Vijver D, Putker K, Engelbeen S, Wijnbergen D, Overzier M, Suidgeest E, van der Weerd L, Aartsma‐Rus A, van Putten M. Learning, memory and blood-brain barrier pathology in Duchenne muscular dystrophy mice lacking Dp427, or Dp427 and Dp140. GENES, BRAIN, AND BEHAVIOR 2024; 23:e12895. [PMID: 38837620 PMCID: PMC11151035 DOI: 10.1111/gbb.12895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/22/2024] [Accepted: 03/28/2024] [Indexed: 06/07/2024]
Abstract
Duchenne muscular dystrophy is a severe neuromuscular disorder that is caused by mutations in the DMD gene, resulting in a disruption of dystrophin production. Next to dystrophin expression in the muscle, different isoforms of the protein are also expressed in the brain and lack of these isoforms leads to cognitive and behavioral deficits in patients. It remains unclear how the loss of the shorter dystrophin isoform Dp140 affects these processes. Using a variety of behavioral tests, we found that mdx and mdx4cv mice (which lack Dp427 or Dp427 + Dp140, respectively) exhibit similar deficits in working memory, movement patterns and blood-brain barrier integrity. Neither model showed deficits in spatial learning and memory, learning flexibility, anxiety or spontaneous behavior, nor did we observe differences in aquaporin 4 and glial fibrillary acidic protein. These results indicate that in contrast to Dp427, Dp140 does not play a crucial role in processes of learning, memory and spontaneous behavior.
Collapse
Affiliation(s)
- Minou Verhaeg
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Kevin Adamzek
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Davy van de Vijver
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Kayleigh Putker
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Sarah Engelbeen
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Daphne Wijnbergen
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Maurice Overzier
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Ernst Suidgeest
- C.J. Gorter MRI Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Louise van der Weerd
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
- C.J. Gorter MRI Center, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Maaike van Putten
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
10
|
Miano-Burkhardt A, Alvarez Jerez P, Daida K, Bandres Ciga S, Billingsley KJ. The Role of Structural Variants in the Genetic Architecture of Parkinson's Disease. Int J Mol Sci 2024; 25:4801. [PMID: 38732020 PMCID: PMC11084710 DOI: 10.3390/ijms25094801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Parkinson's disease (PD) significantly impacts millions of individuals worldwide. Although our understanding of the genetic foundations of PD has advanced, a substantial portion of the genetic variation contributing to disease risk remains unknown. Current PD genetic studies have primarily focused on one form of genetic variation, single nucleotide variants (SNVs), while other important forms of genetic variation, such as structural variants (SVs), are mostly ignored due to the complexity of detecting these variants with traditional sequencing methods. Yet, these forms of genetic variation play crucial roles in gene expression and regulation in the human brain and are causative of numerous neurological disorders, including forms of PD. This review aims to provide a comprehensive overview of our current understanding of the involvement of coding and noncoding SVs in the genetic architecture of PD.
Collapse
Affiliation(s)
- Abigail Miano-Burkhardt
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA; (A.M.-B.); (K.D.)
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD 20892, USA; (P.A.J.); (S.B.C.)
| | - Pilar Alvarez Jerez
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD 20892, USA; (P.A.J.); (S.B.C.)
| | - Kensuke Daida
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA; (A.M.-B.); (K.D.)
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD 20892, USA; (P.A.J.); (S.B.C.)
| | - Sara Bandres Ciga
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD 20892, USA; (P.A.J.); (S.B.C.)
| | - Kimberley J. Billingsley
- Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD 20892, USA; (A.M.-B.); (K.D.)
- Center for Alzheimer’s and Related Dementias, National Institute on Aging, Bethesda, MD 20892, USA; (P.A.J.); (S.B.C.)
| |
Collapse
|
11
|
Childs AM, Turner C, Astin R, Bianchi S, Bourke J, Cunningham V, Edel L, Edwards C, Farrant P, Heraghty J, James M, Massey C, Messer B, Michel Sodhi J, Murphy PB, Schiava M, Thomas A, Trucco F, Guglieri M. Development of respiratory care guidelines for Duchenne muscular dystrophy in the UK: key recommendations for clinical practice. Thorax 2024; 79:476-485. [PMID: 38123347 DOI: 10.1136/thorax-2023-220811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/17/2023] [Indexed: 12/23/2023]
Abstract
Significant inconsistencies in respiratory care provision for Duchenne muscular dystrophy (DMD) are reported across different specialist neuromuscular centres in the UK. The absence of robust clinical evidence and expert consensus is a barrier to the implementation of care recommendations in public healthcare systems as is the need to increase awareness of key aspects of care for those living with DMD. Here, we provide evidenced-based and/or consensus-based best practice for the respiratory care of children and adults living with DMD in the UK, both as part of routine care and in an emergency. METHODOLOGY Initiated by an expert working group of UK-based respiratory physicians (including British Thoracic Society (BTS) representatives), neuromuscular clinicians, physiotherapist and patient representatives, draft guidelines were created based on published evidence, current practice and expert opinion. After wider consultation with UK respiratory teams and neuromuscular services, consensus was achieved on these best practice recommendations for respiratory care in DMD. RESULT The resulting recommendations are presented in the form of a flow chart for assessment and monitoring, with additional guidance and a separate chart setting out key considerations for emergency management. The recommendations have been endorsed by the BTS. CONCLUSIONS These guidelines provide practical, reasoned recommendations for all those managing day-to-day and acute respiratory care in children and adults with DMD. The hope is that this will support patients and healthcare professionals in accessing high standards of care across the UK.
Collapse
Affiliation(s)
- Anne-Marie Childs
- Department of Paediatric Neurosciences, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Catherine Turner
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Ronan Astin
- Division of Medical Specialties, University College London Hospitals NHS Foundation Trust, London, UK
| | - Stephen Bianchi
- Academic Department of Respiratory Medicine, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - John Bourke
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Lisa Edel
- Respiratory Neuromuscular Physiotherapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Christopher Edwards
- Leeds Centre for Children's Respiratory Medicine, Leeds Children's Hospital, Leeds General Infirmary, Leeds, UK
| | | | - Jane Heraghty
- Department of Paediatrics, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Meredith James
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Charlotte Massey
- Queen Square Centre for Neuromuscular Diseases, University College London NHS Foundation Trust, London, UK
- Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Ben Messer
- North East Assisted Ventilation Service, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jassi Michel Sodhi
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Patrick Brian Murphy
- Lane Fox Respiratory Unit, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Marianela Schiava
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ajit Thomas
- Department of Respiratory Medicine, Royal Stoke University Hospital, Stoke-on-Trent, UK
| | - Federica Trucco
- Dubowitz Neuromuscular Centre, University College London, London, UK
- Paediatric Respiratory Medicine, Royal Brompton Hospital, London, UK
| | - Michela Guglieri
- John Walton Muscular Dystrophy Research Centre, Newcastle University Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
12
|
Peruzzo D, Ciceri T, Mascheretti S, Lampis V, Arrigoni F, Agarwal N, Giubergia A, Villa FM, Crippa A, Nobile M, Mani E, Russo A, D'Angelo MG. Brain Alteration Patterns in Children with Duchenne Muscular Dystrophy: A Machine Learning Approach to Magnetic Resonance Imaging. J Neuromuscul Dis 2024:JND230075. [PMID: 38578898 DOI: 10.3233/jnd-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Background Duchenne Muscular Dystrophy (DMD) is a genetic disease in which lack of the dystrophin protein causes progressive muscular weakness, cardiomyopathy and respiratory insufficiency. DMD is often associated with other cognitive and behavioral impairments, however the correlation of abnormal dystrophin expression in the central nervous system with brain structure and functioning remains still unclear. Objective To investigate brain involvement in patients with DMD through a multimodal and multivariate approach accounting for potential comorbidities. Methods We acquired T1-weighted and Diffusion Tensor Imaging data from 18 patients with DMD and 18 age- and sex-matched controls with similar cognitive and behavioral profiles. Cortical thickness, structure volume, fractional anisotropy and mean diffusivity measures were used in a multivariate analysis performed using a Support Vector Machine classifier accounting for potential comorbidities in patients and controls. Results the classification experiment significantly discriminates between the two populations (97.2% accuracy) and the forward model weights showed that DMD mostly affects the microstructural integrity of long fiber bundles, in particular in the cerebellar peduncles (bilaterally), in the posterior thalamic radiation (bilaterally), in the fornix and in the medial lemniscus (bilaterally). We also reported a reduced cortical thickness, mainly in the motor cortex, cingulate cortex, hippocampal area and insula. Conclusions Our study identified a small pattern of alterations in the CNS likely associated with the DMD diagnosis.
Collapse
Affiliation(s)
- Denis Peruzzo
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Tommaso Ciceri
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Sara Mascheretti
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia (PV), Italy
| | - Valentina Lampis
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia (PV), Italy
| | - Filippo Arrigoni
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
- Paediatric Radiology and Neuroradiology Department, V. Buzzi Children's Hospital, Milan, Italy
| | - Nivedita Agarwal
- Diagnostic Imaging and Neuroradiology Unit, Scientific Institute IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Alice Giubergia
- Neuroimaging Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Filippo Maria Villa
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Alessandro Crippa
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Maria Nobile
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Elisa Mani
- Child Psychopathology Unit,Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Annamaria Russo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| | - Maria Grazia D'Angelo
- Unit of Rehabilitation of Rare Diseases of the Central and Peripheral Nervous System, Scientific Institute, IRCCS Eugenio Medea, Bosisio Parini, Italy
| |
Collapse
|
13
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
14
|
Gatto F, Benemei S, Piluso G, Bello L. The complex landscape of DMD mutations: moving towards personalized medicine. Front Genet 2024; 15:1360224. [PMID: 38596212 PMCID: PMC11002111 DOI: 10.3389/fgene.2024.1360224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/26/2024] [Indexed: 04/11/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe genetic disorder characterized by progressive muscle degeneration, with respiratory and cardiac complications, caused by mutations in the DMD gene, encoding the protein dystrophin. Various DMD mutations result in different phenotypes and disease severity. Understanding genotype/phenotype correlations is essential to optimize clinical care, as mutation-specific therapies and innovative therapeutic approaches are becoming available. Disease modifier genes, trans-active variants influencing disease severity and phenotypic expressivity, may modulate the response to therapy, and become new therapeutic targets. Uncovering more disease modifier genes via extensive genomic mapping studies offers the potential to fine-tune prognostic assessments for individuals with DMD. This review provides insights into genotype/phenotype correlations and the influence of modifier genes in DMD.
Collapse
Affiliation(s)
| | | | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Napoli, Italy
| | - Luca Bello
- Department of Neurosciences DNS, University of Padova, Padova, Italy
| |
Collapse
|
15
|
Dori A, Scutifero M, Passamano L, Zoppi D, Ruggiero L, Trabacca A, Politano L. Treatment with ataluren in four symptomatic Duchenne carriers. A pilot study. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2024; 43:8-15. [PMID: 38586166 PMCID: PMC10997039 DOI: 10.36185/2532-1900-398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/01/2024] [Indexed: 04/09/2024]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked neuromuscular disorder caused by dystrophin gene deletions (75%), duplications (15-20%) and point mutations (5-10%), a small portion of which are nonsense mutations. Women carrying dystrophin gene mutations are commonly unaffected because the wild X allele may produce a sufficient amount of the dystrophin protein. However, approximately 8-10% of them may experience muscle symptoms and 50% of those over 40 years develop cardiomyopathy. The presence of symptoms defines the individual as an affected "symptomatic or manifesting carrier". Though there is no effective cure for DMD, therapies are available to slow the decline of muscle strength and delay the onset and progression of cardiac and respiratory impairment. These include ataluren for patients with nonsense mutations, and antisense oligonucleotides therapies, for patients with specific deletions. Symptomatic DMD female carriers are not included in these indications and little data documenting their management, often entrusted to the discretion of individual doctors, is present in the literature. In this article, we report the clinical and instrumental outcomes of four symptomatic DMD carriers, aged between 26 and 45 years, who were treated with ataluren for 21 to 73 months (average 47.3), and annually evaluated for muscle strength, respiratory and cardiological function. Two patients retain independent ambulation at ages 33 and 45, respectively. None of them developed respiratory involvement or cardiomyopathy. No clinical adverse effects or relevant abnormalities in routine laboratory values, were observed.
Collapse
Affiliation(s)
- Amir Dori
- Department of Neurology, Chaim Sheba Medical Center, HaShomer, and Joseph Sagol Neuroscience Center, Faculty of Medicine, Aviv University, Aviv, Israel
| | - Marianna Scutifero
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Luigia Passamano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Dario Zoppi
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Naples, Italy
| | - Lucia Ruggiero
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Naples, Italy
| | - Antonio Trabacca
- Scientific Institute IRCCS “E. Medea”, Unit for Severe disabilities in developmental age and young adults (Developmental Neurology and Neurorehabilitation), Brindisi, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
16
|
Muntoni F, Byrne BJ, McMillan HJ, Ryan MM, Wong BL, Dukart J, Bansal A, Cosson V, Dreghici R, Guridi M, Rabbia M, Staunton H, Tirucherai GS, Yen K, Yuan X, Wagner KR. The Clinical Development of Taldefgrobep Alfa: An Anti-Myostatin Adnectin for the Treatment of Duchenne Muscular Dystrophy. Neurol Ther 2024; 13:183-219. [PMID: 38190001 PMCID: PMC10787703 DOI: 10.1007/s40120-023-00570-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a genetic muscle disorder that manifests during early childhood and is ultimately fatal. Recently approved treatments targeting the genetic cause of DMD are limited to specific subpopulations of patients, highlighting the need for therapies with wider applications. Pharmacologic inhibition of myostatin, an endogenous inhibitor of muscle growth produced almost exclusively in skeletal muscle, has been shown to increase muscle mass in several species, including humans. Taldefgrobep alfa is an anti-myostatin recombinant protein engineered to bind to and block myostatin signaling. Preclinical studies of taldefgrobep alfa demonstrated significant decreases in myostatin and increased lower limb volume in three animal species, including dystrophic mice. METHODS This manuscript reports the cumulative data from three separate clinical trials of taldefgrobep alfa in DMD: a phase 1 study in healthy adult volunteers (NCT02145234), and two randomized, double-blind, placebo-controlled studies in ambulatory boys with DMD-a phase 1b/2 trial assessing safety (NCT02515669) and a phase 2/3 trial including the North Star Ambulatory Assessment (NSAA) as the primary endpoint (NCT03039686). RESULTS In healthy adult volunteers, taldefgrobep alfa was generally well tolerated and resulted in a significant increase in thigh muscle volume. Treatment with taldefgrobep alfa was associated with robust dose-dependent suppression of free myostatin. In the phase 1b/2 trial, myostatin suppression was associated with a positive effect on lean body mass, though effects on muscle mass were modest. The phase 2/3 trial found that the effects of treatment did not meet the primary endpoint pre-specified futility analysis threshold (change from baseline of ≥ 1.5 points on the NSAA total score). CONCLUSIONS The futility analysis demonstrated that taldefgrobep alfa did not result in functional change for boys with DMD. The program was subsequently terminated in 2019. Overall, there were no safety concerns, and no patients were withdrawn from treatment as a result of treatment-related adverse events or serious adverse events. TRIAL REGISTRATION NCT02145234, NCT02515669, NCT03039686.
Collapse
Affiliation(s)
- Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK
- NIHR Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, London, UK
| | | | - Hugh J McMillan
- Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa, ON, Canada
| | - Monique M Ryan
- Royal Children's Hospital, University of Melbourne, Murdoch Children's Research Institute, Melbourne, Australia
| | - Brenda L Wong
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain and Behaviour (INM-7), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | | | - Roxana Dreghici
- F. Hoffmann-La Roche Ltd, Basel, Switzerland
- Solid Biosciences Inc., Cambridge, MA, USA
| | | | | | | | | | - Karl Yen
- Genentech Inc., South San Francisco, CA, USA
- Sanofi, Paris, France
| | | | - Kathryn R Wagner
- F. Hoffmann-La Roche Ltd, Basel, Switzerland.
- The Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
17
|
Liu G, Lipari P, Mollin A, Jung S, Teplova I, Li W, Ying L, More V, Lennox W, Yeh S, McGann E, Moon YC, Rice C, Huarte E, Gruszka B, Ray B, Goodwin E, Buckendahl P, Yurkow E, Braughton B, Narasimhan J, Welch E, Voronin G, Weetall M. Comparison of pharmaceutical properties and biological activities of prednisolone, deflazacort, and vamorolone in DMD disease models. Hum Mol Genet 2024; 33:211-223. [PMID: 37819629 PMCID: PMC10800023 DOI: 10.1093/hmg/ddad173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 09/26/2023] [Accepted: 10/09/2023] [Indexed: 10/13/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disabling X-linked recessive disorder that causes gradual and irreversible loss of muscle, resulting in early death. The corticosteroids prednisone/prednisolone and deflazacort are used to treat DMD as the standard of care; however, only deflazacort is FDA approved for DMD. The novel atypical corticosteroid vamorolone is being investigated for treatment of DMD. We compared the pharmaceutical properties as well as the efficacy and safety of the three corticosteroids across multiple doses in the B10-mdx DMD mouse model. Pharmacokinetic studies in the mouse and evaluation of p-glycoprotein (P-gP) efflux in a cellular system demonstrated that vamorolone is not a strong P-gp substrate resulting in measurable central nervous system (CNS) exposure in the mouse. In contrast, deflazacort and prednisolone are strong P-gp substrates. All three corticosteroids showed efficacy, but also side effects at efficacious doses. After dosing mdx mice for two weeks, all three corticosteroids induced changes in gene expression in the liver and the muscle, but prednisolone and vamorolone induced more changes in the brain than did deflazacort. Both prednisolone and vamorolone induced depression-like behavior. All three corticosteroids reduced endogenous corticosterone levels, increased glucose levels, and reduced osteocalcin levels. Using micro-computed tomography, femur bone density was decreased, reaching significance with prednisolone. The results of these studies indicate that efficacious doses of vamorolone, are associated with similar side effects as seen with other corticosteroids. Further, because vamorolone is not a strong P-gp substrate, vamorolone distributes into the CNS increasing the potential CNS side-effects.
Collapse
Affiliation(s)
- Grace Liu
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Philip Lipari
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Anna Mollin
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Stephen Jung
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Irina Teplova
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Wencheng Li
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Lanqing Ying
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Vijay More
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - William Lennox
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Shirley Yeh
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Eric McGann
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Young-Choon Moon
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Cari Rice
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Eduardo Huarte
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Barbara Gruszka
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Balmiki Ray
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Elizabeth Goodwin
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Patricia Buckendahl
- Rutgers University, Molecular Imaging Center, 41 Gordon Road, Piscataway, NJ 08854, United States
| | - Edward Yurkow
- Rutgers University, Molecular Imaging Center, 41 Gordon Road, Piscataway, NJ 08854, United States
| | - Bruce Braughton
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Jana Narasimhan
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Ellen Welch
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Gregory Voronin
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| | - Marla Weetall
- PTC Therapeutics, Inc., 100 Corporate Court, South Plainfield, NJ 07080, United States
| |
Collapse
|
18
|
Wijekoon N, Gonawala L, Ratnayake P, Liyanage R, Amaratunga D, Hathout Y, Steinbusch HWM, Dalal A, Hoffman EP, de Silva KRD. Title-molecular diagnostics of dystrophinopathies in Sri Lanka towards phenotype predictions: an insight from a South Asian resource limited setting. Eur J Med Res 2024; 29:37. [PMID: 38195599 PMCID: PMC10775540 DOI: 10.1186/s40001-023-01600-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/15/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The phenotype of Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) patients is determined by the type of DMD gene variation, its location, effect on reading frame, and its size. The primary objective of this investigation was to determine the frequency and distribution of DMD gene variants (deletions/duplications) in Sri Lanka through the utilization of a combined approach involving multiplex polymerase chain reaction (mPCR) followed by Multiplex Ligation Dependent Probe Amplification (MLPA) and compare to the international literature. The current consensus is that MLPA is a labor efficient yet expensive technique for identifying deletions and duplications in the DMD gene. METHODOLOGY Genetic analysis was performed in a cohort of 236 clinically suspected pediatric and adult myopathy patients in Sri Lanka, using mPCR and MLPA. A comparative analysis was conducted between our findings and literature data. RESULTS In the entire patient cohort (n = 236), mPCR solely was able to identify deletions in the DMD gene in 131/236 patients (DMD-120, BMD-11). In the same cohort, MLPA confirmed deletions in 149/236 patients [DMD-138, BMD -11]. These findings suggest that mPCR has a detection rate of 95% (131/138) among all patients who received a diagnosis. The distal and proximal deletion hotspots for DMD were exons 45-55 and 6-15. Exon 45-60 identified as a novel in-frame variation hotspot. Exon 45-59 was a hotspot for BMD deletions. Comparisons with the international literature show significant variations observed in deletion and duplication frequencies in DMD gene across different populations. CONCLUSION DMD gene deletions and duplications are concentrated in exons 45-55 and 2-20 respectively, which match global variation hotspots. Disparities in deletion and duplication frequencies were observed when comparing our data to other Asian and Western populations. Identified a 95% deletion detection rate for mPCR, making it a viable initial molecular diagnostic approach for low-resource countries where MLPA could be used to evaluate negative mPCR cases and cases with ambiguous mutation borders. Our findings may have important implications in the early identification of DMD with limited resources in Sri Lanka and to develop tailored molecular diagnostic algorithms that are regional and population specific and easily implemented in resource limited settings.
Collapse
Affiliation(s)
- Nalaka Wijekoon
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200, Maastricht, The Netherlands
| | - Lakmal Gonawala
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200, Maastricht, The Netherlands
| | | | - Roshan Liyanage
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | | | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, 13902, USA
| | - Harry W M Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200, Maastricht, The Netherlands
| | - Ashwin Dalal
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, 500039, India
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, 13902, USA
| | - K Ranil D de Silva
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka.
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200, Maastricht, The Netherlands.
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana, 10390, Sri Lanka.
| |
Collapse
|
19
|
Cope H, Fischer R, Heslop E, McNiff M, Johnson A, Camino E, Denger B, Armstrong N, Thakrar S, Bateman-House A, Beaverson KL, Woollacott IOC, Phillips D, Fernandez V, Ganot A, Donisa-Dreghici R, Mansfield C, Peay H. Clinician Perspectives of Gene Therapy as a Treatment Option for Duchenne Muscular Dystrophy. J Neuromuscul Dis 2024; 11:1085-1093. [PMID: 39093077 PMCID: PMC11380263 DOI: 10.3233/jnd-240033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) is a progressive, life-limiting, neuromuscular disorder. Clinicians play an important role in informing families about therapy options, including approved gene therapies and clinical trials of unapproved therapies. Objective This study aimed to understand the perspectives of clinicians about gene therapy for DMD, which has not previously been studied. Methods We conducted interviews with specialist clinicians treating patients with DMD in the United States (n = 8) and United Kingdom (n = 8). Interviews were completed in 2022, before any approved gene therapies, to gain insight into barriers and facilitators to implementing gene therapy and educational needs of clinicians. Results Most respondents expressed cautious optimism about gene therapy. Responses varied regarding potential benefits with most expecting delayed progression and duration of benefit (1 year to lifelong). Concern about anticipated risks also varied; types of anticipated risks included immunological reactions, liver toxicity, and cardiac or renal dysfunction. Clinicians generally, but not uniformly, understood that gene therapy for DMD would not be curative. Most reported needing demonstrable clinical benefit to justify treatment-related risks. Conclusions Our data demonstrate variability in knowledge and attitudes about gene therapy among clinicians who follow patients with DMD. As our knowledge base about DMD gene therapy grows, clinician education is vital to ensuring that accurate information is communicated to patients and families.
Collapse
Affiliation(s)
- Heidi Cope
- RTI Health Solutions, Research Triangle Park, NC, USA
| | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, D.C., USA
| | - Emma Heslop
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle Upon Tyne, UK
| | - Megan McNiff
- John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, D.C., USA
| | - Brian Denger
- Parent Project Muscular Dystrophy, Washington, D.C., USA
| | - Niki Armstrong
- Parent Project Muscular Dystrophy, Washington, D.C., USA
| | | | - Alison Bateman-House
- Department of Population Health, Division of Medical Ethics, NYU Grossman School of Medicine, New York, NY, USA
| | | | | | | | | | - Annie Ganot
- Solid Biosciences Inc., Charlestown, MA, USA
| | | | | | - Holly Peay
- RTI Health Solutions, Research Triangle Park, NC, USA
| |
Collapse
|
20
|
McDonald C, Camino E, Escandon R, Finkel RS, Fischer R, Flanigan K, Furlong P, Juhasz R, Martin AS, Villa C, Sweeney HL. Draft Guidance for Industry Duchenne Muscular Dystrophy, Becker Muscular Dystrophy, and Related Dystrophinopathies - Developing Potential Treatments for the Entire Spectrum of Disease. J Neuromuscul Dis 2024; 11:499-523. [PMID: 38363616 DOI: 10.3233/jnd-230219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Duchenne muscular dystrophy (DMD) and related dystrophinopathies are neuromuscular conditions with great unmet medical needs that require the development of effective medical treatments. Objective To aid sponsors in clinical development of drugs and therapeutic biological products for treating DMD across the disease spectrum by integrating advancements, patient registries, natural history studies, and more into a comprehensive guidance. Methods This guidance emerged from collaboration between the FDA, the Duchenne community, and industry stakeholders. It entailed a structured approach, involving multiple committees and boards. From its inception in 2014, the guidance underwent revisions incorporating insights from gene therapy studies, cardiac function research, and innovative clinical trial designs. Results The guidance provides a deeper understanding of DMD and its variants, focusing on patient engagement, diagnostic criteria, natural history, biomarkers, and clinical trials. It underscores patient-focused drug development, the significance of dystrophin as a biomarker, and the pivotal role of magnetic resonance imaging in assessing disease progression. Additionally, the guidance addresses cardiomyopathy's prominence in DMD and the burgeoning field of gene therapy. Conclusions The updated guidance offers a comprehensive understanding of DMD, emphasizing patient-centric approaches, innovative trial designs, and the importance of biomarkers. The focus on cardiomyopathy and gene therapy signifies the evolving realm of DMD research. It acts as a crucial roadmap for sponsors, potentially leading to improved treatments for DMD.
Collapse
Affiliation(s)
| | - Eric Camino
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rafael Escandon
- DGBI Consulting, LLC, Bainbridge Island, Washington, DC, USA
| | | | - Ryan Fischer
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Kevin Flanigan
- Center for Experimental Neurotherapeutics, Department of Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Pat Furlong
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Rose Juhasz
- Nationwide Children's Hospital, Columbus, OH, USA
| | - Ann S Martin
- Parent Project Muscular Dystrophy, Washington, DC, USA
| | - Chet Villa
- Trinity Health Michigan, Grand Rapids, MI, USA
| | - H Lee Sweeney
- Cincinnati Children's Hospital Medical Center within the UC Department of Pediatrics, Cincinnati, OH, USA
| |
Collapse
|
21
|
Torices L, Nunes-Xavier CE, Mingo J, Luna S, Erramuzpe A, Cortés JM, Pulido R. Induction of Translational Readthrough on Protein Tyrosine Phosphatases Targeted by Premature Termination Codon Mutations in Human Disease. Methods Mol Biol 2024; 2743:1-19. [PMID: 38147205 DOI: 10.1007/978-1-0716-3569-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Nonsense mutations generating premature termination codons (PTCs) in various genes are frequently associated with somatic cancer and hereditary human diseases since PTCs commonly generate truncated proteins with defective or altered function. Induced translational readthrough during protein biosynthesis facilitates the incorporation of an amino acid at the position of a PTC, allowing the synthesis of a complete protein. This may evade the pathological effect of the PTC mutation and provide new therapeutic opportunities. Several protein tyrosine phosphatases (PTPs) genes are targeted by PTC in human disease, the tumor suppressor PTEN being the more prominent paradigm. Here, using PTEN and laforin as examples, two PTPs from the dual-specificity phosphatase subfamily, we describe methodologies to analyze in silico the distribution and frequency of pathogenic PTC in PTP genes. We also summarize laboratory protocols and technical notes to study the induced translational readthrough reconstitution of the synthesis of PTP targeted by PTC in association with disease in cellular models.
Collapse
Affiliation(s)
- Leire Torices
- Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Caroline E Nunes-Xavier
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Janire Mingo
- Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Sandra Luna
- Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Asier Erramuzpe
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
| | - Jesús M Cortés
- Biobizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain
- Cell Biology and Histology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Rafael Pulido
- Biobizkaia Health Research Institute, Barakaldo, Spain.
- Ikerbasque, The Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
22
|
Chen Y, Ling C, Chen M, Yu L, Yang J, Fang Q. Astaxanthin Ameliorates Worsened Muscle Dysfunction of MDX Mice Fed with a High-Fat Diet through Reducing Lipotoxicity and Regulating Gut Microbiota. Nutrients 2023; 16:33. [PMID: 38201863 PMCID: PMC10780320 DOI: 10.3390/nu16010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Duchenne muscular dystrophy (DMD), a severe X-linked inherited neuromuscular disease, has a high prevalence of obesity. Obesity exacerbates muscle damage and results in adverse clinical outcomes. Preventing obesity helps DMD patients delay disease progression and improve quality of life. Astaxanthin (AX) is a kind of carotenoid which has antioxidant and anti-adipogenesis effects. In this study, male C57BL/10ScSnDmdmdx/J mice were fed with a normal diet, a high-fat diet (HFD), and an HFD containing AX for 16 weeks, respectively. The results showed that AX significantly increased gastrocnemius fiber cross-section area and grip strength, improved treadmill endurance test and mitochondrial morphology, and reduced muscle triglyceride and malonaldehyde levels compared to the HFD. Lipidomic analysis revealed that AX decreased high levels of triglyceride, diglyceride, ceramides, and wax ester induced by HFD. Gut microbiota analysis indicated that AX supplementation failed to alleviate abnormal microbiota diversity, but increased the relative abundances of Akkermansia, Bifidobacterium, Butyricicoccus, and Staphylococcus. In conclusion, AX was expected to alleviate disease progression associated with obesity in DMD patients by reducing lipotoxicity and increasing the abundance of beneficial bacteria.
Collapse
Affiliation(s)
- Ying Chen
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (Y.C.); (L.Y.)
| | - Chenjie Ling
- Department of Clinical Nutrition, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215124, China;
| | - Mengting Chen
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215006, China;
| | - Liqiang Yu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (Y.C.); (L.Y.)
| | - Jing Yang
- Department of Clinical Nutrition, The First Affiliated Hospital of Soochow University, Suzhou 215031, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; (Y.C.); (L.Y.)
- Department of Clinical Nutrition, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215124, China;
| |
Collapse
|
23
|
Sarker S, Eshaque TB, Soorajkumar A, Nassir N, Zehra B, Kanta SI, Rahaman MA, Islam A, Akter S, Ali MK, Mim RA, Uddin KMF, Chowdhury MSJ, Shams N, Baqui MA, Lim ET, Akter H, Woodbury-Smith M, Uddin M. Mutational spectrum and phenotypic variability of Duchenne muscular dystrophy and related disorders in a Bangladeshi population. Sci Rep 2023; 13:21547. [PMID: 38057384 PMCID: PMC10700514 DOI: 10.1038/s41598-023-48982-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe rare neuromuscular disorder caused by mutations in the X-linked dystrophin gene. Several mutations have been identified, yet the full mutational spectrum, and their phenotypic consequences, will require genotyping across different populations. To this end, we undertook the first detailed genotype and phenotype characterization of DMD in the Bangladeshi population. We investigated the rare mutational and phenotypic spectrum of the DMD gene in 36 DMD-suspected Bangladeshi participants using an economically affordable diagnostic strategy involving initial screening for exonic deletions in the DMD gene via multiplex PCR, followed by testing PCR-negative patients for mutations using whole exome sequencing. The deletion mapping identified two critical DMD gene hotspot regions (near proximal and distal ends, spanning exons 8-17 and exons 45-53, respectively) that comprised 95% (21/22) of the deletions for this population cohort. From our exome analysis, we detected two novel pathogenic hemizygous mutations in exons 21 and 42 of the DMD gene, and novel pathogenic recessive and loss of function variants in four additional genes: SGCD, DYSF, COL6A3, and DOK7. Our phenotypic analysis showed that DMD suspected participants presented diverse phenotypes according to the location of the mutation and which gene was impacted. Our study provides ethnicity specific new insights into both clinical and genetic aspects of DMD.
Collapse
Affiliation(s)
- Shaoli Sarker
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
- Bangladesh Shishu Hospital and Institute, Dhaka, Bangladesh
| | | | - Anjana Soorajkumar
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Nasna Nassir
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Binte Zehra
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | | | - Md Atikur Rahaman
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Amirul Islam
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
- GenomeArc Inc., Mississauga, Ontario, Canada
| | - Shimu Akter
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Mohammad Kawsar Ali
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Rabeya Akter Mim
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - K M Furkan Uddin
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | | | - Nusrat Shams
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Md Abdul Baqui
- Department of Biochemistry, Holy Family Red Crescent Medical College and Hospital, Dhaka, Bangladesh
| | - Elaine T Lim
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Hosneara Akter
- Genetics and Genomic Medicine Centre (GGMC), NeuroGen Healthcare, Dhaka, Bangladesh
| | - Marc Woodbury-Smith
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Mohammed Uddin
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE.
- GenomeArc Inc., Mississauga, Ontario, Canada.
| |
Collapse
|
24
|
Singer A, Aartsma-Rus A, Grinshpun-Cohen J, Sagi-Dain L. Lessons learned from the first national population-based genetic carrier-screening program for Duchenne muscular dystrophy. Genet Med 2023; 25:100981. [PMID: 37712502 DOI: 10.1016/j.gim.2023.100981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023] Open
Abstract
PURPOSE To summarize the results of first year implementation of pan-ethnic screening testing for Duchenne muscular dystrophy (DMD) and present the ensuing challenges. METHODS Data acquisition for this study was performed by retrospective search of Ministry of Health registry for reports of all laboratories performing genetic screening tests. DMD testing was performed by multiplex ligation-dependent probe amplification technology. In case of single-exon deletion, sequencing of the specific exon was performed to rule out underlying single-nucleotide variant. RESULTS Of overall 85,737 DMD tests, 82 clinically significant findings were noted (0.095%, or 1:1,046 women). In addition, 80 findings with uncertain clinical significance were detected (0.093%, or 1:1072), as well as 373 cases (0.4%, or 1:230) of single-exon deletions subsequently identified as false positives because of underlying single-nucleotide variant, mostly variants in exon 8 in North African Jewish population, and in exon 48 in Arab Muslim population. CONCLUSION Interpretation of population-based DMD carrier screening is complex, occasionally requiring additional genetic testing methods and ethical considerations. Multicenter data registry, including ethnic origin and familial segregation in selected cases, is crucial for optimal definition of the results during genetic counseling and informed decisions regarding prenatal testing.
Collapse
Affiliation(s)
- Amihood Singer
- Community Genetics, Public Health Services, Ministry of Health, Jerusalem, Israel
| | - Annemieke Aartsma-Rus
- Dutch Center for RNA Therapeutics, The Netherlands; Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Lena Sagi-Dain
- Genetics Institute, Carmel Medical Center, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
25
|
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
|
26
|
Wallach E, Ehlinger V, Biotteau M, Walther-Louvier U, Péréon Y, Vuillerot C, Fontaine S, Sabouraud P, Espil-Taris C, Cuisset JM, Laugel V, Baudou E, Arnaud C, Cances C. Confirmatory validation of the french version of the Duchenne Muscular Dystrophy module of the pediatric quality of life inventory (PedsQL TM3.0DMDfv). BMC Pediatr 2023; 23:563. [PMID: 37968589 PMCID: PMC10647061 DOI: 10.1186/s12887-023-04153-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 06/24/2023] [Indexed: 11/17/2023] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a neuromuscular disease that inevitably leads to total loss of autonomy. The new therapeutic strategies aim to both improve survival and optimise quality of life. Evaluating quality of life is nevertheless a major challenge. No DMD-specific quality of life scale to exists in French. We therefore produced a French translation of the English Duchenne Muscular Dystrophy module of the Pediatric Quality of Life Inventory (PedsQLTMDMD) following international recommendations. The study objective was to carry out a confirmatory validation of the French version of the PedsQLTMDMD for paediatric patients with DMD, using French multicentre descriptive cross-sectional data. The sample consisted of 107 patients. Internal consistency was acceptable for proxy-assessments, with Cronbach's alpha coefficients above 0.70, except for the Treatment dimension. For self-assessments, internal consistency was acceptable only for the Daily Activities dimension. Our results showed poor metric qualities for the French version of the PedsQLTMDMD based on a sample of about 100 children, but these results remained consistent with those of the original validation. This confirms the interest of its use in clinical practice.
Collapse
Affiliation(s)
- Elisabeth Wallach
- Neuropediatric Department, Toulouse-Purpan University Hospital, Toulouse, France.
| | - Virginie Ehlinger
- UMR 1295 CERPOP, Toulouse University, Inserm, University Toulouse III Paul Sabatier, Toulouse, France
| | - Maelle Biotteau
- Neuropediatric Department, Toulouse-Purpan University Hospital, Toulouse, France.
- ToNIC, Toulouse NeuroImaging Center, University of Toulouse, Inserm, UPS, Toulouse, France.
| | - Ulrike Walther-Louvier
- CHU Montpellier, Service de Neuropédiatrie, Centre de Référence Maladies Neuromusculaires AOC, Montpellier, France
| | - Yann Péréon
- Reference Centre for Neuromuscular Diseases AOC, Filnemus, Euro-NMD, Hôtel-Dieu, CHU Nantes, Nantes, France
| | - Carole Vuillerot
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Service de Médecine Physique et de Réadaptation Pédiatrique, Bron, France
- NeuroMyogen Institute, CNRS UMR 5310 - INSERM U1217, University of Lyon, Lyon, France
| | - Stephanie Fontaine
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, L'Escale, Service de Médecine Physique et de Réadaptation Pédiatrique, Bron, France
| | - Pascal Sabouraud
- Department of Paediatrics, French Reference Center for Neuromuscular Diseases, American Memorial Hospital, Reims University Hospital Center, Reims, France
| | - Caroline Espil-Taris
- CHU Pellegrin, Service de neuropédiatrie, Centre de Référence Maladies Neuromusculaires AOC, Bordeaux, France
| | - Jean-Marie Cuisset
- Reference Centre for Neuromuscular Diseases Nord/Est/Ile-de-France, CHU Lille, Lille, France
- Department of Pediatric Neurology, CHU Lille, Lille, France
| | - Vincent Laugel
- Unité de neuropédiatrie et CIC pédiatrique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Eloïse Baudou
- Neuropediatric Department, Toulouse-Purpan University Hospital, Toulouse, France
| | - Catherine Arnaud
- UMR 1295 CERPOP, Toulouse University, Inserm, University Toulouse III Paul Sabatier, Toulouse, France
- Clinical Epidemiology Unit, University Hospital, Toulouse, France
| | - Claude Cances
- Neuropediatric Department, Toulouse-Purpan University Hospital, Toulouse, France
| |
Collapse
|
27
|
Mercuri E, Pane M, Cicala G, Brogna C, Ciafaloni E. Detecting early signs in Duchenne muscular dystrophy: comprehensive review and diagnostic implications. Front Pediatr 2023; 11:1276144. [PMID: 38027286 PMCID: PMC10667703 DOI: 10.3389/fped.2023.1276144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Despite the early onset of clinical signs suggestive of Duchenne muscular dystrophy (DMD), a diagnosis is often not made until four years of age or older, with a diagnostic delay of up to two years from the appearance of the first symptoms. As disease-modifying therapies for DMD become available that are ideally started early before irreversible muscle damage occurs, the importance of avoiding diagnostic delay increases. Shortening the time to a definite diagnosis in DMD allows timely genetic counseling and assessment of carrier status, initiation of multidisciplinary standard care, timely initiation of appropriate treatments, and precise genetic mutation characterization to assess suitability for access to drugs targeted at specific mutations while reducing the emotional and psychological family burden of the disease. This comprehensive literature review describes the early signs of impairment in DMD and highlights the bottlenecks related to the different diagnostic steps. In summary, the evidence suggests that the best mitigation strategy for improving the age at diagnosis is to increase awareness of the early symptoms of DMD and encourage early clinical screening with an inexpensive and sensitive serum creatine kinase test in all boys who present signs of developmental delay and specific motor test abnormality at routine pediatrician visits.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Marika Pane
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianpaolo Cicala
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudia Brogna
- Pediatric Neurology, Università Cattolica del Sacro Cuore, Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Emma Ciafaloni
- Department of Neurology, University of Rochester, Rochester, NY, United States
| |
Collapse
|
28
|
Zaidman CM, Proud CM, McDonald CM, Lehman KJ, Goedeker NL, Mason S, Murphy AP, Guridi M, Wang S, Reid C, Darton E, Wandel C, Lewis S, Malhotra J, Griffin DA, Potter RA, Rodino-Klapac LR, Mendell JR. Delandistrogene Moxeparvovec Gene Therapy in Ambulatory Patients (Aged ≥4 to <8 Years) with Duchenne Muscular Dystrophy: 1-Year Interim Results from Study SRP-9001-103 (ENDEAVOR). Ann Neurol 2023; 94:955-968. [PMID: 37539981 DOI: 10.1002/ana.26755] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/05/2023]
Abstract
OBJECTIVE Delandistrogene moxeparvovec is approved in the USA for the treatment of ambulatory patients (4-5 years) with Duchenne muscular dystrophy. ENDEAVOR (SRP-9001-103; NCT04626674) is a single-arm, open-label study to evaluate delandistrogene moxeparvovec micro-dystrophin expression, safety, and functional outcomes following administration of commercial process delandistrogene moxeparvovec. METHODS In cohort 1 of ENDEAVOR (N = 20), eligible ambulatory males, aged ≥4 to <8 years, received a single intravenous infusion of delandistrogene moxeparvovec (1.33 × 1014 vg/kg). The primary endpoint was change from baseline (CFBL) to week 12 in delandistrogene moxeparvovec micro-dystrophin by western blot. Additional endpoints evaluated included: safety; vector genome copies; CFBL to week 12 in muscle fiber-localized micro-dystrophin by immunofluorescence; and functional assessments, including North Star Ambulatory Assessment, with comparison with a propensity score-weighted external natural history control. RESULTS The 1-year safety profile of commercial process delandistrogene moxeparvovec in ENDEAVOR was consistent with safety data reported in other delandistrogene moxeparvovec trials (NCT03375164 and NCT03769116). Delandistrogene moxeparvovec micro-dystrophin expression was robust, with sarcolemmal localization at week 12; mean (SD) CFBL in western blot, 54.2% (42.6); p < 0.0001. At 1 year, patients demonstrated stabilized or improved North Star Ambulatory Assessment total scores; mean (SD) CFBL, +4.0 (3.5). Treatment versus a propensity score-weighted external natural history control demonstrated a statistically significant difference in least squares mean (standard error) CFBL in North Star Ambulatory Assessment, +3.2 (0.6) points; p < 0.0001. INTERPRETATION Results confirm efficient transduction of muscle by delandistrogene moxeparvovec. One-year post-treatment, delandistrogene moxeparvovec was well tolerated, and demonstrated stabilized or improved motor function, suggesting a clinical benefit for patients with Duchenne muscular dystrophy. ANN NEUROL 2023;94:955-968.
Collapse
Affiliation(s)
- Craig M Zaidman
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Crystal M Proud
- Children's Hospital of the King's Daughters, Norfolk, VA, USA
| | | | - Kelly J Lehman
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
| | - Natalie L Goedeker
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | | | | | | | | | - Carol Reid
- Roche Products Ltd, Welwyn Garden City, UK
| | | | | | - Sarah Lewis
- Sarepta Therapeutics, Inc., Cambridge, MA, USA
| | | | | | | | | | - Jerry R Mendell
- Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH, USA
- The Ohio State University, Columbus, OH, USA
| |
Collapse
|
29
|
Kulkarni A, Tanga S, Karmakar A, Hota A, Maji B. CRISPR-Based Precision Molecular Diagnostics for Disease Detection and Surveillance. ACS APPLIED BIO MATERIALS 2023; 6:3927-3945. [PMID: 37788375 DOI: 10.1021/acsabm.3c00439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Sensitive, rapid, and portable molecular diagnostics is the future of disease surveillance, containment, and therapy. The recent SARS-CoV-2 pandemic has reminded us of the vulnerability of lives from ever-evolving pathogens. At the same time, it has provided opportunities to bridge the gap by translating basic molecular biology into therapeutic tools. One such molecular biology technique is CRISPR (clustered regularly interspaced short palindromic repeat) which has revolutionized the field of molecular diagnostics at the need of the hour. The use of CRISPR-Cas systems has been widespread in biology research due to the ease of performing genetic manipulations. In 2012, CRISPR-Cas systems were, for the first time, shown to be reprogrammable, i.e., capable of performing sequence-specific gene editing. This discovery catapulted the field of CRISPR-Cas research and opened many unexplored avenues in the field of gene editing, from basic research to therapeutics. One such field that benefitted greatly from this discovery was molecular diagnostics, as using CRISPR-Cas technologies enabled existing diagnostic methods to become more sensitive, accurate, and portable, a necessity in disease control. This Review aims to capture some of the trajectories and advances made in this arena and provides a comprehensive understanding of the methods and their potential use as point-of-care diagnostics.
Collapse
Affiliation(s)
- Akshara Kulkarni
- Ashoka University, Department of Biology, Rajiv Gandhi Education City, Sonipat, Haryana 131029, India
| | - Sadiya Tanga
- Ashoka University, Department of Chemistry, Rajiv Gandhi Education City, Sonipat, Haryana 131029, India
| | - Arkadeep Karmakar
- Bose Institute, Department of Biological Sciences, EN Block, Sector V, Kolkata 700091, West Bengal, India
| | - Arpita Hota
- Bose Institute, Department of Biological Sciences, EN Block, Sector V, Kolkata 700091, West Bengal, India
| | - Basudeb Maji
- Ashoka University, Department of Biology, Rajiv Gandhi Education City, Sonipat, Haryana 131029, India
- Ashoka University, Department of Chemistry, Rajiv Gandhi Education City, Sonipat, Haryana 131029, India
- Bose Institute, Department of Biological Sciences, EN Block, Sector V, Kolkata 700091, West Bengal, India
| |
Collapse
|
30
|
Kaspute G, Arunagiri BD, Alexander R, Ramanavicius A, Samukaite-Bubniene U. Development of Essential Oil Delivery Systems by 'Click Chemistry' Methods: Possible Ways to Manage Duchenne Muscular Dystrophy. MATERIALS (BASEL, SWITZERLAND) 2023; 16:6537. [PMID: 37834674 PMCID: PMC10573547 DOI: 10.3390/ma16196537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
Recently, rare diseases have received attention due to the need for improvement in diagnosed patients' and their families' lives. Duchenne muscular dystrophy (DMD) is a rare, severe, progressive, muscle-wasting disease. Today, the therapeutic standard for treating DMD is corticosteroids, which cause serious adverse side effects. Nutraceuticals, e.g., herbal extracts or essential oils (EOs), are possible active substances to develop new drug delivery systems to improve DMD patients' lives. New drug delivery systems lead to new drug effects, improved safety and accuracy, and new therapies for rare diseases. Herbal extracts and EOs combined with click chemistry can lead to the development of safer treatments for DMD. In this review, we focus on the need for novel drug delivery systems using EOs as the therapy for DMD and the potential use of click chemistry for drug delivery systems. New EO complex drug delivery systems may offer a new approach for improving muscle conditions and mental health issues associated with DMD. However, further research should identify the potential of these systems in the context of DMD. In this review, we discuss possibilities for applying EOs to DMD before implementing expensive research in a theoretical way.
Collapse
Affiliation(s)
- Greta Kaspute
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Bharani Dharan Arunagiri
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Rakshana Alexander
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| | - Urte Samukaite-Bubniene
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekis av. 3, LT-10257 Vilnius, Lithuania;
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, LT-03225 Vilnius, Lithuania; (B.D.A.); (R.A.)
| |
Collapse
|
31
|
Magot A, Wahbi K, Leturcq F, Jaffre S, Péréon Y, Sole G. Diagnosis and management of Becker muscular dystrophy: the French guidelines. J Neurol 2023; 270:4763-4781. [PMID: 37422773 DOI: 10.1007/s00415-023-11837-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/11/2023]
Abstract
Becker muscular dystrophy (BMD) is one of the most frequent among neuromuscular diseases, affecting approximately 1 in 18,000 male births. It is linked to a genetic mutation on the X chromosome. In contrast to Duchenne muscular dystrophy, for which improved care and management have changed the prognosis and life expectancy of patients, few guidelines have been published for management of BMD. Many clinicians are inexperienced in managing the complications of this disease. In France, a committee of experts from a wide range of disciplines met in 2019 to establish recommendations, with the goal of improving care of patients with BMD. Here, we present the tools to provide diagnosis of BMD as quickly as possible and for differential diagnoses. Then, we describe the multidisciplinary approach essential for optimum management of BMD. We give recommendations for the initial assessment and follow-up of the neurological, respiratory, cardiac, and orthopedic consequences of males who present with BMD. Finally, we describe the optimal therapeutic management of these complications. We also provide guidance on cardiac management for female carriers.
Collapse
Affiliation(s)
- Armelle Magot
- Centre de Référence des Maladies Neuromusculaires AOC, Laboratoire d'Explorations Fonctionnelles, FILNEMUS, Hôtel-Dieu, CHU de Nantes, Nantes, France.
| | - Karim Wahbi
- Centre de Référence des Maladies Neuromusculaires Paris Est, AP-HP, Hopital Pitie salpêtrière, Institut de myologie, Cardiovascular Research Center, Inserm Unit 970, Paris, France
| | - France Leturcq
- Service de Médecine Génomique des Maladies de Système et d'Organe, APHP, Université de Paris, Hopital Cochin, Paris, France
| | - Sandrine Jaffre
- Service de Pneumologie, L'institut du thorax, Hôpital G et R Laennec, CHU de Nantes, Nantes, France
| | - Yann Péréon
- Centre de Référence des Maladies Neuromusculaires AOC, Laboratoire d'Explorations Fonctionnelles, FILNEMUS, Euro-NMD, Hôtel-Dieu, CHU de Nantes, Nantes, France
| | - Guilhem Sole
- Centre de Référence des Maladies Neuromusculaires AOC, FILNEMUS, Hôpital Pellegrin, CHU de Bordeaux, Bordeaux, France
| |
Collapse
|
32
|
Jayash SN, Hamoudi D, Stephen LA, Argaw A, Huesa C, Joseph S, Wong SC, Frenette J, Farquharson C. Anti-RANKL Therapy Prevents Glucocorticoid-Induced Bone Loss and Promotes Muscle Function in a Mouse Model of Duchenne Muscular Dystrophy. Calcif Tissue Int 2023; 113:449-468. [PMID: 37470794 PMCID: PMC10516841 DOI: 10.1007/s00223-023-01116-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023]
Abstract
Bisphosphonates prevent bone loss in glucocorticoid (GC)-treated boys with Duchenne muscular dystrophy (DMD) and are recommended as standard of care. Targeting receptor activator of nuclear factor kappa-B ligand (RANKL) may have advantages in DMD by ameliorating dystrophic skeletal muscle function in addition to their bone anti-resorptive properties. However, the potential effects of anti-RANKL treatment upon discontinuation in GC-induced animal models of DMD are unknown and need further investigation prior to exploration in the clinical research setting. In the first study, the effects of anti-RANKL and deflazacort (DFZ) on dystrophic skeletal muscle function and bone microstructure were assessed in mdx mice treated with DFZ or anti-RANKL, or both for 8 weeks. Anti-RANKL and DFZ improved grip force performance of mdx mice but an additive effect was not noted. However, anti-RANKL but not DFZ improved ex vivo contractile properties of dystrophic muscles. This functional improvement was associated with a reduction in muscle damage and fibrosis, and inflammatory cell number. Anti-RANKL treatment, with or without DFZ, also improved trabecular bone structure of mdx mice. In a second study, intravenous zoledronate (Zol) administration (1 or 2 doses) following 2 months of discontinuation of anti-RANKL treatment was mostly required to record an improvement in bone microarchitecture and biomechanical properties in DFZ-treated mdx mice. In conclusion, the ability of anti-RANKL therapy to restore muscle function has profound implications for DMD patients as it offers the possibility of improving skeletal muscle function without the steroid-related skeletal side effects.
Collapse
Affiliation(s)
- Soher Nagi Jayash
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Dounia Hamoudi
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Centre, Hospitalier de L’Université Laval, Université Laval, Quebec City, QC Canada
| | - Louise A. Stephen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| | - Anteneh Argaw
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Centre, Hospitalier de L’Université Laval, Université Laval, Quebec City, QC Canada
| | - Carmen Huesa
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Shuko Joseph
- Royal Hospital for Children Glasgow, School of Medicine, Dentistry and Nursing, Child Health, Queen Elizabeth University Hospital, Glasgow, UK
| | - Sze Choong Wong
- University of Glasgow/Royal Hospital for Children Glasgow, School of Medicine, Dentistry & Nursing, Child Health, Queen Elizabeth University Hospital, Glasgow, UK
| | - Jérôme Frenette
- Centre de Recherche du Centre Hospitalier, Universitaire de Québec-Centre, Hospitalier de L’Université Laval, Université Laval, Quebec City, QC Canada
| | - Colin Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian, EH25 9RG UK
| |
Collapse
|
33
|
Agrawal P, Harish V, Mohd S, Singh SK, Tewari D, Tatiparthi R, Harshita, Vishwas S, Sutrapu S, Dua K, Gulati M. Role of CRISPR/Cas9 in the treatment of Duchenne muscular dystrophy and its delivery strategies. Life Sci 2023; 330:122003. [PMID: 37544379 DOI: 10.1016/j.lfs.2023.122003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder brought on by mutations in the DMD gene, which prevent muscle cells from expressing the dystrophin protein. CRISPR/Cas9 technology has evolved as potential option to treat DMD due to its ability to permanently skip exons, restoring the disrupted DMD reading frame and leading to dystrophin restoration. Even though, having potential to treat DMD, the delivery, safety and efficacy of this technology is still challenging. Several delivery methods, including viral vectors, nanoparticles, and electroporation, have been explored to deliver CRISPR/Cas9 to the targeted cells. Despite the potential of CRISPR/Cas9 technology in the treatment of DMD, several limitations need to be addressed. The off-target effects of CRISPR/Cas9 are a major concern that needs to be addressed to avoid unintended mutations. The delivery of CRISPR/Cas9 to the target cells and the immune response due to the viral vectors used for delivery are a few other limitations. The clinical trials of CRISPR/Cas9 for DMD provide valuable insights into the safety and efficacy of this technology in humans and the limitations that need to be known. Therefore, in this review we insightfully discussed the challenges and limitations of CRISPR/Cas9 in the treatment of DMD and delivery strategies used, and the ongoing efforts to overcome these challenges and restore dystrophin expression in DMD patients in the ongoing trials.
Collapse
Affiliation(s)
- Pooja Agrawal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Vancha Harish
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Sharfuddin Mohd
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi 110017, India
| | - Ramanjireddy Tatiparthi
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Harshita
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Srinivas Sutrapu
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia; Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
34
|
Berntsson SG, Matsson H, Kristoffersson A, Niemelä V, van Duyvenvoorde HA, Richel-van Assenbergh C, van der Klift HM, Casar-Borota O, Frykholm C, Landtblom AM. Case report: a novel deep intronic splice-altering variant in DMD as a cause of Becker muscular dystrophy. Front Genet 2023; 14:1226766. [PMID: 37795243 PMCID: PMC10546389 DOI: 10.3389/fgene.2023.1226766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
We present the case of a male patient who was ultimately diagnosed with Becker muscular dystrophy (BMD; MIM# 300376) after the onset of muscle weakness in his teens progressively led to significant walking difficulties in his twenties. A genetic diagnosis was pursued but initial investigation revealed no aberrations in the dystrophin gene (DMD), although immunohistochemistry and Western blot analysis suggested the diagnosis of dystrophinopathy. Eventually, after more than 10 years, an RNA analysis captured abnormal splicing where 154 nucleotides from intron 43 were inserted between exon 43 and 44 resulting in a frameshift and a premature stop codon. Normal splicing of the DMD gene was also observed. Additionally, a novel variant c.6291-13537A>G in DMD was confirmed in the genomic DNA of the patient. The predicted function of the variant aligns with the mRNA results. To conclude, we here demonstrate that mRNA analysis can guide the diagnosis of non-coding genetic variants in DMD.
Collapse
Affiliation(s)
| | - Hans Matsson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Clinical Genetics, Rudbeck Laboratory, Uppsala University Hospital, Uppsala, Sweden
| | - Anna Kristoffersson
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
| | - Valter Niemelä
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
| | | | | | | | - Olivera Casar-Borota
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Clinical Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Carina Frykholm
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Clinical Genetics, Rudbeck Laboratory, Uppsala University Hospital, Uppsala, Sweden
| | - Anne-Marie Landtblom
- Department of Medical Sciences, Neurology, Uppsala University, Uppsala, Sweden
- Department of Clinical Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health, Linköping University, Linköping, Sweden
| |
Collapse
|
35
|
Wu J, Ren L, Huang X, Hu L, Zhang L, Xie D, Li Z, Han N, Huang S. Identification of Two Novel Variants of the DMD Gene in Chinese Families with Duchenne Muscular Dystrophy. Pharmgenomics Pers Med 2023; 16:759-766. [PMID: 37609034 PMCID: PMC10441636 DOI: 10.2147/pgpm.s416294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 08/11/2023] [Indexed: 08/24/2023] Open
Abstract
Background Duchenne muscular dystrophy (DMD), an X-linked recessive neuromuscular disorder, is caused by pathogenic variants in the DMD gene encoding a large structural protein in muscle cells. Methods Two probands, a 6-year old boy and a 1-month old infant, respectively, were clinically diagnosed with DMD based on elevated levels of creatine kinase and creatine kinase isoenzyme. CNVplex and whole exome sequencing (WES) were performed for causal variants, and Sanger sequencing was used for verification. Results CNVplex found no large deletions or duplications in the DMD gene in both patients, but WES discovered a single-nucleotide deletion in exon 48 (NM_004006.2:c.6963del, p.Asp2322ThrfsTer16) in the proband of pedigree 1, and a nonsense mutation in exon 27 (NM_004006.2:c.3637A>T, p.K1213Ter) in the proband of pedigree 2. Conclusion The results of our study expand the mutation spectrum of DMD and enrich our understanding of the clinical characteristics of DMD. Genetic counseling was provided for the two families involved in this study.
Collapse
Affiliation(s)
- Jiangfen Wu
- GuiZhou University Medical College, Guiyang, Guizhou, People’s Republic of China
| | - Lingyan Ren
- Prenatal Diagnosis Center, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
| | - Xinyi Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Li Hu
- Prenatal Diagnosis Center, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
| | - Liangliang Zhang
- Prenatal Diagnosis Center, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
| | - Dan Xie
- GuiZhou University Medical College, Guiyang, Guizhou, People’s Republic of China
| | - Zhimin Li
- Annoroad Gene Technology (Beijing) Co., Ltd, Beijing, People’s Republic of China
| | - Naijian Han
- Annoroad Gene Technology (Beijing) Co., Ltd, Beijing, People’s Republic of China
| | - Shengwen Huang
- GuiZhou University Medical College, Guiyang, Guizhou, People’s Republic of China
- Prenatal Diagnosis Center, Guizhou Provincial People’s Hospital, Guiyang, People’s Republic of China
- NHC Key Laboratory of Pulmonary Immunological Diseases, Guizhou Provincial, People’s Hospital, Guiyang, People’s Republic of China
| |
Collapse
|
36
|
Stirm M, Shashikadze B, Blutke A, Kemter E, Lange A, Stöckl JB, Jaudas F, Laane L, Kurome M, Keßler B, Zakhartchenko V, Bähr A, Klymiuk N, Nagashima H, Walter MC, Wurst W, Kupatt C, Fröhlich T, Wolf E. Systemic deletion of DMD exon 51 rescues clinically severe Duchenne muscular dystrophy in a pig model lacking DMD exon 52. Proc Natl Acad Sci U S A 2023; 120:e2301250120. [PMID: 37428903 PMCID: PMC10629550 DOI: 10.1073/pnas.2301250120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 06/10/2023] [Indexed: 07/12/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked disease caused by mutations in the DMD gene, leading to complete absence of dystrophin and progressive degeneration of skeletal musculature and myocardium. In DMD patients and in a corresponding pig model with a deletion of DMD exon 52 (DMDΔ52), expression of an internally shortened dystrophin can be achieved by skipping of DMD exon 51 to reframe the transcript. To predict the best possible outcome of this strategy, we generated DMDΔ51-52 pigs, additionally representing a model for Becker muscular dystrophy (BMD). DMDΔ51-52 skeletal muscle and myocardium samples stained positive for dystrophin and did not show the characteristic dystrophic alterations observed in DMDΔ52 pigs. Western blot analysis confirmed the presence of dystrophin in the skeletal muscle and myocardium of DMDΔ51-52 pigs and its absence in DMDΔ52 pigs. The proteome profile of skeletal muscle, which showed a large number of abundance alterations in DMDΔ52 vs. wild-type (WT) samples, was normalized in DMDΔ51-52 samples. Cardiac function at age 3.5 mo was significantly reduced in DMDΔ52 pigs (mean left ventricular ejection fraction 58.8% vs. 70.3% in WT) but completely rescued in DMDΔ51-52 pigs (72.3%), in line with normalization of the myocardial proteome profile. Our findings indicate that ubiquitous deletion of DMD exon 51 in DMDΔ52 pigs largely rescues the rapidly progressing, severe muscular dystrophy and the reduced cardiac function of this model. Long-term follow-up studies of DMDΔ51-52 pigs will show if they develop symptoms of the milder BMD.
Collapse
Affiliation(s)
- Michael Stirm
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Bachuki Shashikadze
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich81377, Germany
| | - Andreas Blutke
- Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU Munich, Munich80539, Germany
| | - Elisabeth Kemter
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Andreas Lange
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Jan B. Stöckl
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich81377, Germany
| | - Florian Jaudas
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Laeticia Laane
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Mayuko Kurome
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Barbara Keßler
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Valeri Zakhartchenko
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
| | - Andrea Bähr
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich81675, Germany
| | - Nikolai Klymiuk
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich81675, Germany
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, Kawasaki214-8571, Japan
| | - Maggie C. Walter
- Department of Neurology, Friedrich Baur Institute, LMU Munich, Munich80336, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Munich, Neuherberg85674, Germany
- Chair of Developmental Genetics, TUM School of Life Sciences, Technische Universität München, Freising85354, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich81675, Germany
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich81675, Germany
| | - Thomas Fröhlich
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich81377, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, Munich81377, Germany
- Center for Innovative Medical Models, Department of Veterinary Sciences, LMU Munich, Oberschleissheim85764, Germany
- Laboratory for Functional Genome Analysis, Gene Center, LMU Munich, Munich81377, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer, LMU Munich, Munich81377, Germany
| |
Collapse
|
37
|
Czifrus E, Berlau DJ. Viltolarsen: a treatment option for Duchenne muscular dystrophy patients who are amenable to exon 53 skipping therapy. Expert Rev Neurother 2023; 23:853-858. [PMID: 37572081 DOI: 10.1080/14737175.2023.2246658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/07/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) is a progressive genetic disease leading to muscular weakness. DMD is caused by mutations of the dystrophin gene on the X chromosome that is responsible for production of dystrophin protein. Dystrophin contributes to structural support in muscle cells and mutations result in dystrophin protein deficiency which causes muscle damage and the associated clinical presentation. Exon skipping medications, including the exon 53 targeting viltolarsen, are the first agents with the ability to partially restore dystrophin protein. AREAS COVERED Herein, the authors profile viltolarsen for the DMD patients who are amenable to exon 53 skipping therapy and provide their expert perspectives on this subject. EXPERT OPINION Current findings suggest that viltolarsen could play a role in the current and possible future treatment of DMD. Viltolarsen seems to be safe and restores dystrophin protein to around 6% of the normal level. Due to orphan drug status, after the completion of the phase 2 clinical trial, viltolarsen was granted accelerated approval in Japan and in the US. A phase 3 trial is currently in progress and needs to earn full approval. Although a multidisciplinary approach continues to be critical, the addition of exon skipping agents like viltolarsen may improve the quality of patients' lives. However, data on the long-term safety and efficacy of this medication are not yet available due to its recent accelerated approval.
Collapse
Affiliation(s)
- Eszter Czifrus
- Semmelweis University Faculty of Medicine, Budapest, Hungary
| | - Daniel J Berlau
- Department of Pharmaceutical Sciences, Regis University School of Pharmacy, Denver, CO, USA
| |
Collapse
|
38
|
Umurzakova A, Ayaganov D, Mannapova A, Dzhumasheva B, Dzhaksybayeva A. Preliminary Results of a Genetic Study of Children with Duchenne Myodystrophy in Aktobe, Kazakhstan. ARCHIVES OF RAZI INSTITUTE 2023; 78:949-954. [PMID: 38028863 PMCID: PMC10657926 DOI: 10.22092/ari.2022.360050.2534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 10/18/2022] [Indexed: 12/01/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive neuromuscular illness with a progressive course caused by mutations in the gene encoding the protein dystrophin (DMD; locus Xp21. 2). This study aimed to investigate the clinical aspects of DMD progression according to the mutation type. Included in the study were 38 boys aged 3 to 11 years. Laboratory (biochemical evaluation of the level of creatinine phosphokinase, multiplex ligation-dependent probe amplification [MLPA], and next-generation sequencing [NGS] analysis of the DMD gene), genealogical, and clinical approaches were utilized (including adapted Hammersmith Functional Motor Scale, the study of auditory-speech memory by the "Memorizing 10 words" method, and a general neurological assessment). The MLPA revealed deletion in 22 cases (57.8%), duplication in 6 (15.7%), and negative results in 11 (26.5%). To discover point mutations, 11 infants with negative MLPA results were sequenced. According to the results of the NGS, point mutations were identified in six boys (four single-nucleotide deletions and two single-nucleotide duplications), and five boys lacked mutations. Due to the high proportion of neuro-hereditary diseases in the general structure of neurological pathology, the profound disability of patients with progressive mental and physical disadaptation, as well as the generally fatal course of these incurable afflictions, molecular genetics research is of particular importance.
Collapse
Affiliation(s)
- A Umurzakova
- West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - D Ayaganov
- West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - A Mannapova
- West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - B Dzhumasheva
- Regional Children's Clinical Hospital, Aktobe, Kazakhstan
| | | |
Collapse
|
39
|
Onódi Z, Szabó PL, Kucsera D, Pokreisz P, Dostal C, Hilber K, Oudit GY, Podesser BK, Ferdinandy P, Varga ZV, Kiss A. Inflammasome Activity in the Skeletal Muscle and Heart of Rodent Models for Duchenne Muscular Dystrophy. Int J Mol Sci 2023; 24:8497. [PMID: 37239853 PMCID: PMC10218525 DOI: 10.3390/ijms24108497] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by wasting of muscles that leads to difficulty moving and premature death, mainly from heart failure. Glucocorticoids are applied in the management of the disease, supporting the hypothesis that inflammation may be driver as well as target. However, the inflammatory mechanisms during progression of cardiac and skeletal muscle dysfunction are still not well characterized. Our objective was to characterize the inflammasomes in myocardial and skeletal muscle in rodent models of DMD. Gastrocnemius and heart samples were collected from mdx mice and DMDmdx rats (3 and 9-10 months). Inflammasome sensors and effectors were assessed by immunoblotting. Histology was used to assess leukocyte infiltration and fibrosis. In gastrocnemius, a tendency towards elevation of gasdermin D irrespective of the age of the animal was observed. The adaptor protein was elevated in the mdx mouse skeletal muscle and heart. Increased cleavage of the cytokines was observed in the skeletal muscle of the DMDmdx rats. Sensor or cytokine expression was not changed in the tissue samples of the mdx mice. In conclusion, inflammatory responses are distinct between the skeletal muscle and heart in relevant models of DMD. Inflammation tends to decrease over time, supporting the clinical observations that the efficacy of anti-inflammatory therapies might be more prominent in the early stage.
Collapse
Affiliation(s)
- Zsófia Onódi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Petra Lujza Szabó
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Péter Pokreisz
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Christopher Dostal
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Karlheinz Hilber
- Department of Neurophysiology & Neuropharmacology, Center for Physiology & Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Bruno K. Podesser
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- Pharmahungary Group, 6728 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Z.O.)
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, 1085 Budapest, Hungary
| | - Attila Kiss
- Ludwig Boltzmann Institute for Cardiovascular Research at the Center for Biomedical Research and Translational Surgery, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
40
|
Chen CC, Liao RY, Yeh FY, Lin YR, Wu TY, Pastor AE, Zul DD, Hsu YC, Wu KY, Liu KF, Kannagi R, Chen JY, Cai BH. A Simple and Affordable Method to Create Nonsense Mutation Clones of p53 for Studying the Premature Termination Codon Readthrough Activity of PTC124. Biomedicines 2023; 11:biomedicines11051310. [PMID: 37238980 DOI: 10.3390/biomedicines11051310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: A premature termination codon (PTC) can be induced by a type of point mutation known as a nonsense mutation, which occurs within the coding region. Approximately 3.8% of human cancer patients have nonsense mutations of p53. However, the non-aminoglycoside drug PTC124 has shown potential to promote PTC readthrough and rescue full-length proteins. The COSMIC database contains 201 types of p53 nonsense mutations in cancers. We built a simple and affordable method to create different nonsense mutation clones of p53 for the study of the PTC readthrough activity of PTC124. (2) Methods: A modified inverse PCR-based site-directed mutagenesis method was used to clone the four nonsense mutations of p53, including W91X, S94X, R306X, and R342X. Each clone was transfected into p53 null H1299 cells and then treated with 50 μM of PTC124. (3) Results: PTC124 induced p53 re-expression in H1299-R306X and H1299-R342X clones but not in H1299-W91X and H1299-S94X clones. (4) Conclusions: Our data showed that PTC124 more effectively rescued the C-terminal of p53 nonsense mutations than the N-terminal of p53 nonsense mutations. We introduced a fast and low-cost site-directed mutagenesis method to clone the different nonsense mutations of p53 for drug screening.
Collapse
Affiliation(s)
- Chia-Chi Chen
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Physical Therapy, I-Shou University, Kaohsiung City 82445, Taiwan
- School of Chinese Medicine for Post Baccalaureate, I-Shou University, Kaohsiung City 82445, Taiwan
- Department of Pathology, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Ruo-Yu Liao
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Fang-Yu Yeh
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Yu-Rou Lin
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Tze-You Wu
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Alexa Escobar Pastor
- School of Medicine for International Students, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Danny Danilo Zul
- School of Medicine for International Students, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Yun-Chien Hsu
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Kuan-Yo Wu
- Department of Biological Science and Technology, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ke-Fang Liu
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Reiji Kannagi
- Institute of Biomedical Sciences, Academia Sinica, Taipei City 11529, Taiwan
| | - Jang-Yi Chen
- Institute of Biology and Anatomy, National Defense Medical Center, Taipei City 11529, Taiwan
| | - Bi-He Cai
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| |
Collapse
|
41
|
Codina A, Roldán M, Natera-de Benito D, Ortez C, Planas R, Matalonga L, Cuadras D, Carrera L, Exposito J, Marquez J, Jimenez-Mallebrera C, M. Porta J, Nascimento A, Jou C. Innovative Computerized Dystrophin Quantification Method Based on Spectral Confocal Microscopy. Int J Mol Sci 2023; 24:ijms24076358. [PMID: 37047330 PMCID: PMC10094132 DOI: 10.3390/ijms24076358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/16/2022] [Accepted: 12/29/2022] [Indexed: 03/30/2023] Open
Abstract
Several clinical trials are working on drug development for Duchenne and Becker muscular dystrophy (DMD and BMD) treatment, and, since the expected increase in dystrophin is relatively subtle, high-sensitivity quantification methods are necessary. There is also a need to quantify dystrophin to reach a definitive diagnosis in individuals with mild BMD, and in female carriers. We developed a method for the quantification of dystrophin in DMD and BMD patients using spectral confocal microscopy. It offers the possibility to capture the whole emission spectrum for any antibody, ensuring the selection of the emission peak and allowing the detection of fluorescent emissions of very low intensities. Fluorescence was evaluated first on manually selected regions of interest (ROIs), proving the usefulness of the methodology. Later, ROI selection was automated to make it operator-independent. The proposed methodology correctly classified patients according to their diagnosis, detected even minimal traces of dystrophin, and the results obtained automatically were statistically comparable to the manual ones. Thus, spectral imaging could be implemented to measure dystrophin expression and it could pave the way for detailed analysis of how its expression relates to the clinical course. Studies could be further expanded to better understand the expression of dystrophin-associated protein complexes (DAPCs).
Collapse
|
42
|
Wang P, Li H, Zhu M, Han RY, Guo S, Han R. Correction of DMD in human iPSC-derived cardiomyocytes by base-editing-induced exon skipping. Mol Ther Methods Clin Dev 2023; 28:40-50. [PMID: 36588820 PMCID: PMC9792405 DOI: 10.1016/j.omtm.2022.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
Duchenne muscular dystrophy (DMD) is caused by mutations in the DMD gene. Previously, we showed that adenine base editing (ABE) can efficiently correct a nonsense point mutation in a DMD mouse model. Here, we explored the feasibility of base-editing-mediated exon skipping as a therapeutic strategy for DMD using cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs). We first generated a DMD hiPSC line with a large deletion spanning exon 48 through 54 (ΔE48-54) using CRISPR-Cas9 gene editing. Dystrophin expression was disrupted in DMD hiPSC-derived cardiomyocytes (iCMs) as examined by RT-PCR, western blot, and immunofluorescence staining. Transfection of ABE and a guide RNA (gRNA) targeting the splice acceptor led to efficient conversion of AG to GG (35.9% ± 5.7%) and enabled exon 55 skipping. Complete AG to GG conversion in a single clone restored dystrophin expression (42.5% ± 11% of wild type [WT]) in DMD iCMs. Moreover, we designed gRNAs to target the splice sites of exons 6, 7, 8, 43, 44, 46, and 53 in the mutational hotspots and demonstrated their efficiency to induce exon skipping in iCMs. These results highlight the great promise of ABE-mediated exon skipping as a promising therapeutic approach for DMD.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Haiwen Li
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mandi Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Rena Y. Han
- Olentangy Liberty High School, Powell, OH 43065, USA
| | - Shuliang Guo
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
43
|
Li G, Jin M, Li Z, Xiao Q, Lin J, Yang D, Liu Y, Wang X, Xie L, Ying W, Wang H, Zuo E, Shi L, Wang N, Chen W, Xu C, Yang H. Mini-dCas13X-mediated RNA editing restores dystrophin expression in a humanized mouse model of Duchenne muscular dystrophy. J Clin Invest 2023; 133:162809. [PMID: 36512423 PMCID: PMC9888377 DOI: 10.1172/jci162809] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Approximately 10% of monogenic diseases are caused by nonsense point mutations that generate premature termination codons (PTCs), resulting in a truncated protein and nonsense-mediated decay of the mutant mRNAs. Here, we demonstrate a mini-dCas13X-mediated RNA adenine base editing (mxABE) strategy to treat nonsense mutation-related monogenic diseases via A-to-G editing in a genetically humanized mouse model of Duchenne muscular dystrophy (DMD). Initially, we identified a nonsense point mutation (c.4174C>T, p.Gln1392*) in the DMD gene of a patient and validated its pathogenicity in humanized mice. In this model, mxABE packaged in a single adeno-associated virus (AAV) reached A-to-G editing rates up to 84% in vivo, at least 20-fold greater than rates reported in previous studies using other RNA editing modalities. Furthermore, mxABE restored robust expression of dystrophin protein to over 50% of WT levels by enabling PTC read-through in multiple muscle tissues. Importantly, systemic delivery of mxABE by AAV also rescued dystrophin expression to averages of 37%, 6%, and 54% of WT levels in the diaphragm, tibialis anterior, and heart muscle, respectively, as well as rescued muscle function. Our data strongly suggest that mxABE-based strategies may be a viable new treatment modality for DMD and other monogenic diseases.
Collapse
Affiliation(s)
- Guoling Li
- HuiGene Therapeutics, Shanghai, China.,Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Ming Jin
- Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | | | - Qingquan Xiao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiajia Lin
- Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dong Yang
- HuiGene Therapeutics, Shanghai, China.,Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yuanhua Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xing Wang
- HuiGene Therapeutics, Shanghai, China
| | - Long Xie
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Wenqin Ying
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | | | - Erwei Zuo
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Linyu Shi
- HuiGene Therapeutics, Shanghai, China
| | - Ning Wang
- Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Wanjin Chen
- Department of Neurology, First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Chunlong Xu
- Lingang Laboratory, Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| | - Hui Yang
- HuiGene Therapeutics, Shanghai, China.,Institute of Neuroscience, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai, China
| |
Collapse
|
44
|
Quak ZX, Tan SML, Tan KB, Lin W, Chai P, Ng KWP. A manifesting female carrier of Duchenne muscular dystrophy: Importance of genetics for the dystrophinopathies. Singapore Med J 2023; 64:81-87. [PMID: 36722521 PMCID: PMC9979793 DOI: 10.4103/singaporemedj.smj-2021-356] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Zhi Xuan Quak
- Division of Neurology, Department of Medicine, National University Hospital, Singapore
| | | | - Kong Bing Tan
- Yong Loo Lin School of Medicine, National University of Singapore,Department of Pathology, National University Hospital, Singapore
| | - Weiqin Lin
- Yong Loo Lin School of Medicine, National University of Singapore,Department of Cardiology, National University Heart Centre, Singapore
| | - Ping Chai
- Yong Loo Lin School of Medicine, National University of Singapore,Department of Cardiology, National University Heart Centre, Singapore
| | - Kay Wei Ping Ng
- Division of Neurology, Department of Medicine, National University Hospital, Singapore,Yong Loo Lin School of Medicine, National University of Singapore,Correspondence: Dr Kay Wei Ping Ng, Division of Neurology, Department of Medicine, National University Hospital, 1E Kent Ridge Road, 119228, Singapore. E-mail:
| |
Collapse
|
45
|
Viggiano E, Picillo E, Passamano L, Onore ME, Piluso G, Scutifero M, Torella A, Nigro V, Politano L. Spectrum of Genetic Variants in the Dystrophin Gene: A Single Centre Retrospective Analysis of 750 Duchenne and Becker Patients from Southern Italy. Genes (Basel) 2023; 14:214. [PMID: 36672955 PMCID: PMC9859256 DOI: 10.3390/genes14010214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/29/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Dystrophinopathies are X-linked recessive muscle disorders caused by mutations in the dystrophin (DMD) gene that include deletions, duplications, and point mutations. Correct diagnosis is important for providing adequate patient care and family planning, especially at this time when mutation-specific therapies are available. We report a large single-centre study on the spectrum of DMD gene variants observed in 750 patients analyzed for suspected Duchenne (DMD) or Becker (BMD) muscular dystrophy, over the past 30 years, at the Cardiomyology and Medical Genetics of the University of Campania. We found 534 (71.21%) large deletions, 73 (9.73%) large duplications, and 112 (14.93%) point mutations, of which 44 (5.9%) were small ins/del causing frame-shifts, 57 (7.6%) nonsense mutations, 8 (1.1%) splice site and 3 (0.4%) intronic mutations, and 31 (4.13%) non mutations. Moreover, we report the prevalence of the different types of mutations in patients with DMD and BMD according to their decade of birth, from 1930 to 2020, and correlate the data to the different techniques used over the years. In the most recent decades, we observed an apparent increase in the prevalence of point mutations, probably due to the use of Next-Generation Sequencing (NGS). In conclusion, in southern Italy, deletions are the most frequent variation observed in DMD and BMD patients followed by point mutations and duplications, as elsewhere in the world. NGS was useful to identify point mutations in cases of strong suspicion of DMD/BMD negative on deletions/duplications analyses. In the era of personalized medicine and availability of new causative therapies, a collective effort is necessary to enable DMD and BMD patients to have timely genetic diagnoses and avoid late implementation of standard of care and late initiation of appropriate treatment.
Collapse
Affiliation(s)
- Emanuela Viggiano
- Department of Prevention, Hygiene and Public Health Service, ASL Roma 2, 00157 Rome, Italy
| | - Esther Picillo
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Luigia Passamano
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Maria Elena Onore
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Giulio Piluso
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Marianna Scutifero
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Annalaura Torella
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| | - Vincenzo Nigro
- Medical Genetics and Cardiomyology, Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
- Telethon Institute of Genetics and Medicine, 80078 Pozzuoli, Italy
| | - Luisa Politano
- Cardiomyology and Medical Genetics, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Napoli, Italy
| |
Collapse
|
46
|
Xu J, Suo L, Zhao J, Cai T, Mei N, Du P, Gao C, Fang Y, Jiang Y, Zhang JA. MBL2 polymorphism may be a protective factor of autoimmune thyroid disease susceptibility. Mol Genet Genomics 2023; 298:95-105. [PMID: 36318338 DOI: 10.1007/s00438-022-01960-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/28/2022] [Indexed: 01/07/2023]
Abstract
Genetic susceptibility is an essential pathogenetic mechanism in autoimmune thyroid disease (AITD). MBL2 gene polymorphisms have been shown to play a vital role in the pathogenesis of multiple autoimmune disorders, but its contribution to AITD is unclear. The aim of this study was to assess the linkage between MBL2 gene polymorphisms and AITD susceptibility in a Chinese Han population. One thousand seven hundred sixty seven subjects consisting of 965 AITD patients and 802 controls from a Chinese Han population were enrolled in the case-control study. Four common single-nucleotide polymorphisms (SNPs) in the MBL2 gene were tested using high-throughput sequencing technology for sequence-based SNP genotyping. The allele and genotype distribution results showed that the minor alleles of rs198266, rs10824793, and rs4935046 were significantly lower in Hashimoto's thyroiditis (HT) patients than in healthy controls. In further genetic model analysis, the dominant models of rs1982266, rs10824793, and rs4935046 for MBL2 in the AITD group exhibited a lower risk of morbidity. Finally, we discovered that haplotype AAGC was associated with Graves' disease (GD), while AGC was associated with HT. Our study provides strong evidence for a genetic correlation between MBL2 and AITD, and the polymorphism of the MBL2 gene may be a protective factor for AITD, especially for HT. These findings can advance our understanding of the etiology of AITD, as well as provide guidance for prevention and intervention toward AITD.
Collapse
Affiliation(s)
- Jianbin Xu
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Lixia Suo
- Department of Endocrinology, Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai, 201899, China
| | - Jing Zhao
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Tiantian Cai
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Na Mei
- Department of Rehabilitation, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Peng Du
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Chaoqun Gao
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Yudie Fang
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China
| | - Yanfei Jiang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China.
| | - Jin-An Zhang
- Department of Endocrinology and Rheumatology, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, No. 1500 Zhouyuan Road, Pudong District, Shanghai, 201318, China.
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
47
|
SATTENAPALLI NC, ARETI AR, KOTESWARA RAO SN, ALAVALA RR, KULANDAIVELU US. Prevalence Study of Duchene Muscular Dystrophy and its Genetic Sequence in Southern India. IRANIAN JOURNAL OF CHILD NEUROLOGY 2023; 17:29-37. [PMID: 36721834 PMCID: PMC9881832 DOI: 10.22037/ijcn.v17i2.35071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 05/28/2022] [Indexed: 02/02/2023]
Abstract
Objective Duchene Muscular dystrophy (DMD) is the common X-linked heterogenous progressive muscular dystrophy characterized by mutations in the DMD gene. The frequency of dystrophin gene mutations is varied in different DMD population. A precise diagnosis can help to reduce the severity of DMD since it aids in planning of targeted medical treatment and required therapies. This study was aimed to investigate the mutation type, their rate and distribution of DMD'S in southern India. Materials & Materials An observational study was conducted on 250 genetically confirmed DMD patients from March,2019 to March,2021. The distribution pattern and rate of mutations (deletion, duplication, nonsense mutations, minor mutations) were investigated. Results Mutation spectrum was studied on 250 DMD patients, of which 63% exon deletion pattern were reported. 16% deletions were detected in proximal hot region (exons 3-28). The duplications were found 21% in the proximal hotspot largest region (exon 3-25). 16% of the patients reported single deletion (45 exon), 10.7% reported deletions of exon 44. Point mutations detected in 6%, small mutations were detected in 1.2%, non-sense mutations were detected in 2% of study population respectively. Missense Mutations were detected in 0.8% of study population. Conclusion This study estimates mutation spectrum of exon deletion pattern (63%) was predominantly identified in distal region; duplication was most frequent in proximal region. Point mutations, Nonsense mutations and small mutations have a least accountability. This study adds a real world evidence for developing research therapies in DMD.
Collapse
Affiliation(s)
| | - Anka Rao ARETI
- K L College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram-522502, Guntur, AP, India
| | - Siva Naga KOTESWARA RAO
- K L College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram-522502, Guntur, AP, India
| | - Rajasekhar Reddy ALAVALA
- K L College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram-522502, Guntur, AP, India
| | - Uma Sankar KULANDAIVELU
- K L College of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram-522502, Guntur, AP, India
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW This article reviews the history, epidemiology, genetics, clinical presentation, multidisciplinary management, and established and emerging therapies for the dystrophinopathies. RECENT FINDINGS The multidisciplinary care of individuals with dystrophinopathies continues to improve in many ways, including early surveillance and implementation of respiratory, cardiac, and orthopedic health management. The era of genetic therapeutics has altered the treatment landscape in neuromuscular disorders, including the dystrophinopathies. SUMMARY The dystrophinopathies are a spectrum of X-linked genetic disorders characterized by childhood-onset progressive weakness and variable cardiac and cognitive involvement. Corticosteroids are the mainstay of therapy to slow disease progression. Additional strategies for disease amelioration and dystrophin restoration, including gene replacement therapy, are under investigation.
Collapse
|
49
|
Ohtani H, Saotome M, Sakamoto A, Suwa K, Maekawa Y. Drug-refractory Heart Failure in Female Carrier of Duchenne Muscular Dystrophy: A Case of X-linked Dilated Cardiomyopathy. Intern Med 2022. [PMID: 36450469 PMCID: PMC10400384 DOI: 10.2169/internalmedicine.0745-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
A 56-year-old woman was referred to our hospital for the further evaluation of drug-refractory heart failure with a reduced ejection fraction. A family history interview revealed that men in her family had died of Duchenne muscular dystrophy (DMD), whereas she had no skeletal muscle disorder. Myocardial histopathology revealed a reduced dystrophin expression in the cardiomyocyte membrane, and a dystrophin (DMD) gene analysis identified a duplication in exon 8-9 on Xp21, suggesting that she had a cardiac-specific phenotype of dystrophinopathy, i.e. X-linked dilated cardiomyopathy (XLDCM). In conclusion, careful family history interviews and an investigation of dystrophinopathy are required to detect XLDCM in women.
Collapse
Affiliation(s)
- Hayato Ohtani
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Masao Saotome
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Atsushi Sakamoto
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Kenichiro Suwa
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| | - Yuichiro Maekawa
- Department of Cardiology, Internal Medicine 3, Hamamatsu University School of Medicine, Japan
| |
Collapse
|
50
|
Dp71 and intellectual disability in Indonesian patients with Duchenne muscular dystrophy. PLoS One 2022; 17:e0276640. [PMID: 36315559 PMCID: PMC9621454 DOI: 10.1371/journal.pone.0276640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 10/10/2022] [Indexed: 11/12/2022] Open
Abstract
INTRODUCTIONS Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive muscular disease marked by developmental delays due to mutations in the DMD gene, which encodes dystrophin. Brain comorbidity adds to the burden of limited mobility and significantly impacts patients' quality of life and their family. The changes of expression of dystrophin isoforms in the brain due to DMD gene mutations are thought to be related to the cognitive and neurobehavior profiles of DMD. OBJECTIVES This cross-sectional study aimed to characterize cognitive and neurodevelopmental profiles of patients with DMD and to explore underlying genotype-phenotype associations. METHODS Patients with DMD aged 5-18 years from Dr Sardjito Hospital and Universitas Gadjah Mada Academic Hospital from 2017-2022 were included. Multiplex ligation-dependent probe amplification and whole exome sequencing were used to determine mutations in the DMD genes. Cognitive function was measured by intelligence quotient testing using the Wechsler Intelligence Scale for Children and adaptive function tests with Vineland Adaptive Behavior Scales. The Autism Mental Status Exam and Abbreviated Conner's Rating Scale were used to screen for autism spectrum disorder (ASD) and attention deficit and hyperactivity disorder (ADHD), respectively. RESULTS The mean total IQ score of DMD patients was lower than that of the general population (80.6 ± 22.0 vs 100 ± 15), with intellectual disability observed in 15 boys (29.4%). Of the 51 patients with DMD, the Dp71 group had the lowest cognitive performance with a total IQ score (46 ± 24.8; p = 0.003), while the Dp427 group and Dp140 group's total IQ scores were 83.0 ± 24.6 and 84.2 ± 17.5 respectively. There were no DMD patients with ASD, while 4 boys (7.8%) had comorbidity with ADHD. CONCLUSION Boys with DMD are at higher risk of intellectual disability. The risk appears to increase with mutations at the 3' end of the gene (Dp71 disruption). Moreover, Dp71 disruption might not be associated with ADHD and ASD in patients with DMD.
Collapse
|