1
|
Fujimoto T, Mori M, Tonosaki M, Yaoi T, Nakano K, Okamura T, Itoh K. Characterization of Dystrophin Dp71 Expression and Interaction Partners in Embryonic Brain Development: Implications for Duchenne/Becker Muscular Dystrophy. Mol Neurobiol 2025; 62:6256-6272. [PMID: 39760982 DOI: 10.1007/s12035-024-04676-6] [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: 06/08/2024] [Accepted: 12/17/2024] [Indexed: 01/07/2025]
Abstract
Duchenne/Becker muscular dystrophy (DMD/BMD) manifests progressive muscular dystrophy and non-progressive central nervous disorder. The neural disorder is possibly caused by abnormalities in the developmental period; however, basic research to understand the mechanisms remains underdeveloped. The responsible gene, Dmd (dystrophin), generates multiple products derived from several gene promoters. Here, we aim to characterize the expression of the shortest product, Dp71, during embryonic brain development and to identify its interaction proteins by using Dp71-specific tag-insertion mice. We showed that Dp71 and Dp140 were major dystrophin products significantly detectable in the mouse embryonic brains and Dp71 was the only dystrophin product derived from intron-62 gene promoter in the physiological mouse brains. Although both Dp71f (exon 78-exclusive form) and Dp71d (exon 78-inclusive form) existed in the embryonic brains, Dp71f and Dp71d were dominant forms in the prenatal and postnatal periods, respectively. We histologically found that Dp71 was prominently expressed in the neuroepithelium of the dorsal and medial telencephalon, which gives rise to the primordial cerebral cortex and hippocampus. Deeper analysis using in vitro primary culture verified Dp71 expressions in Nestin-positive neural stem/progenitor, Fabp7-positive radial glia, and Gfap-positive astrocytic cell populations. Interestingly, Dp71 was downregulated upon neuronal differentiation from stem/progenitor cells into TuJ1-positive immature neurons; however, Dp71 became detectable at Gephyrin-positive inhibitory postsynapses within mature neurons. Importantly, interactome analysis revealed dystroglycan, dystrobrevins, and syntrophins as dominant Dp71-partners in the embryonic neural stem/progenitor cells. Thus, the presence of Dp71-dystroglycan macromolecular complex was clearly established at an early stage of embryonic brain development, which sheds light on relations between fetal abnormalities and intellectual disabilities in DMD/BMD.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-Cho, Kawaramachi Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| | - Miyuki Mori
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-Cho, Kawaramachi Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Madoka Tonosaki
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-Cho, Kawaramachi Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Takeshi Yaoi
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-Cho, Kawaramachi Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, 162-8655, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, 162-8655, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-Cho, Kawaramachi Hirokoji, Kamigyo-Ku, Kyoto, 602-8566, Japan
| |
Collapse
|
2
|
Sarvutiene J, Ramanavicius A, Ramanavicius S, Prentice U. Advances in Duchenne Muscular Dystrophy: Diagnostic Techniques and Dystrophin Domain Insights. Int J Mol Sci 2025; 26:3579. [PMID: 40332074 PMCID: PMC12027135 DOI: 10.3390/ijms26083579] [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: 02/11/2025] [Revised: 03/27/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Abnormalities in X chromosomes, either numerical or structural, cause X-linked disorders, such as Duchenne muscular dystrophy (DMD). Recent molecular and cytogenetic techniques can help identify DMD gene mutations. The accurate diagnosis of Duchenne is crucial, directly impacting patient treatment management, genetics, and the establishment of effective prevention strategies. This review provides an overview of X chromosomal disorders affecting Duchenne and discusses how mutations in Dystrophin domains can impact detection accuracy. Firstly, the efficiency and use of cytogenetic and molecular techniques for the genetic diagnosis of Duchenne disease have, thus, become increasingly important. Secondly, artificial intelligence (AI) will be instrumental in developing future therapies by enabling the aggregation and synthesis of extensive and heterogeneous datasets, thereby elucidating underlying molecular mechanisms. However, despite advances in diagnostic technology, understanding the role of Dystrophin in Duchenne disease remains a challenge. Therefore, this review aims to synthesize this complex information to significantly advance the understanding of DMD and how it could affect patient care.
Collapse
Affiliation(s)
- Julija Sarvutiene
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
| | - Arunas Ramanavicius
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko St. 24, LT-03225 Vilnius, Lithuania
| | - Simonas Ramanavicius
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
| | - Urte Prentice
- State Research Institute Center for Physical Sciences and Technology (FTMC), Sauletekio Av. 3, LT-10257 Vilnius, Lithuania; (J.S.); (A.R.); (S.R.)
- Department of Physical Chemistry, Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko St. 24, LT-03225 Vilnius, Lithuania
- Department of Personalised Medicine, State Research Institute Center for Innovative Medicine, Santariskiu St. 5, LT-08410 Vilnius, Lithuania
| |
Collapse
|
3
|
Lupu M, Marcu MA, Epure DA, Vladacenco OA, Severin EM, Teleanu RI. Lost in Transition: Challenges in the Journey from Pediatric to Adult Care for a Romanian DMD Patient. Healthcare (Basel) 2025; 13:830. [PMID: 40218127 PMCID: PMC11988853 DOI: 10.3390/healthcare13070830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND The transition from pediatric to adult care in Duchenne Muscular Dystrophy (DMD) is challenging due to the disease's complexity and the need for lifelong, comprehensive management. In Romania, ongoing efforts aim to enhance multidisciplinary collaboration, though systemic barriers, such as fragmented healthcare services, persist. Nonsense mutations, including those in exon 30 described here, are often associated with more severe disease progression. METHODS We present the case of a 17-year-old Romanian DMD patient with a nonsense mutation in exon 30 of the dystrophin gene. The patient received multidisciplinary pediatric care addressing his medical needs, including neuromuscular, respiratory, cardiac, and orthopedic management. Transition readiness was assessed using the Transition Readiness Assessment Questionnaire (TRAQ), and the patient's perspective on the process was documented. RESULTS Care followed international standards, but the disease progressed predictably, with gradual loss of ambulation, respiratory decline, and cardiac complications. The TRAQ revealed strengths in communication with healthcare providers but moderate confidence in self-management tasks. From the patient's perspective, fragmented adult services and difficulty accessing specialized neuromuscular support remain major obstacles, underscoring the importance of early, structured transition planning and patient-centered approaches. CONCLUSIONS Transitioning to adult services requires strong communication between pediatric and adult teams and integration of specialized care. Tailored follow-up plans ensure continuity of care and effective disease management. This case reflects broader needs in similar healthcare contexts, highlighting the necessity of robust transition frameworks to respond to patient-specific challenges and ultimately support long-term quality of life.
Collapse
Affiliation(s)
- Maria Lupu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Maria-Alexandra Marcu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Diana Anamaria Epure
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| | - Oana Aurelia Vladacenco
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| | - Emilia Maria Severin
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
| | - Raluca Ioana Teleanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania; (M.L.); (O.A.V.); (E.M.S.); (R.I.T.)
- Department of Paediatric Neurology, Dr Victor Gomoiu Children’s Hospital, 022102 Bucharest, Romania;
| |
Collapse
|
4
|
Tetorou K, Aghaeipour A, Singh S, Morgan JE, Muntoni F. The role of dystrophin isoforms and interactors in the brain. Brain 2025; 148:1081-1098. [PMID: 39673425 PMCID: PMC11967788 DOI: 10.1093/brain/awae384] [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: 04/24/2024] [Revised: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 12/16/2024] Open
Abstract
Dystrophin is a protein crucial for maintaining the structural integrity of skeletal muscle. So far, attention has been focused on the role of dystrophin in muscle, in view of the devastating progression of weakness and early death that characterizes Duchenne muscular dystrophy. However, in the last few years, the role of shorter dystrophin isoforms, including development and adult expression-specific mechanisms, has been a greater focus. Within the cerebral landscape, various cell types, such as glia, oligodendrocytes and Purkinje, cerebellar granule and vascular-associated cells express a spectrum of dystrophin isoforms, including Dp427, Dp140, Dp71 and Dp40. The interaction of these isoforms with a multitude of proteins suggests their involvement in neurotransmission, influencing several circuit functions. This review presents the intricate interactions among dystrophin isoforms and diverse protein complexes across different cell types and brain regions, as well as the associated clinical complications. We focus on studies investigating protein interactions with dystrophin in the past 30 years at a biochemical level. In essence, the brain's dystrophin landscape is a thrilling exploration of diversity, challenging preconceptions and opening new avenues for understanding CNS physiology. It also holds potential therapeutic implications for neurological complications involving brain dystrophin deficiency. By revealing the molecular complexities related to dystrophin, this review paves the way for future investigations and therapeutic interventions for this CNS aspect of Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Konstantina Tetorou
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Artadokht Aghaeipour
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Simran Singh
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Jennifer E Morgan
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| | - Francesco Muntoni
- Developmental Neurosciences Department, Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Developmental Neurosciences Department, National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
5
|
Kenepp A, Russell-Giller S, Seehra S, Fee R, Hinton VJ. Reading skills over time among children with Duchenne muscular dystrophy. Child Neuropsychol 2025; 31:467-486. [PMID: 39088241 PMCID: PMC11785819 DOI: 10.1080/09297049.2024.2386078] [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: 12/14/2023] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked progressive neuromuscular disorder with a distinct cognitive profile including decreased verbal span. Children with DMD are also at risk for lower scores on academic achievement tests and increased behavioral problems. Longitudinal analyses generally reveal a stable intellectual profile, although attention and behavioral problems may negatively impact longitudinal IQ scores. To date, no study has reported on reading over time in DMD. Reading performance was assessed longitudinally in children with DMD, examining for potential contributions to the trajectory. Retrospective data analysis on assessments completed at baseline, year 2, and year 4 on 26 boys with DMD and 27 unaffected sibling controls (age at baseline: DMD 8 ± 1.4, controls 9 ± 2.6) indicated that children with DMD performed slightly, yet significantly, worse than controls on reading skills, but the longitudinal trajectory of reading skills for children with DMD and controls was not significantly different. Verbal span at time 1 was uniquely associated with later reading skills in children with DMD. Behavior was not associated with declines. The results confirm that children with DMD underperform on reading tasks and align with previous research suggesting that cognitive skills in DMD are stable over time.
Collapse
Affiliation(s)
- Amanda Kenepp
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Shira Russell-Giller
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Sonia Seehra
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| | - Robert Fee
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Veronica J Hinton
- Queens College, City University of New York, Flushing, NY, USA
- The Graduate Center, City University of New York, New York, NY, USA
| |
Collapse
|
6
|
Jones L, Divakar S, Collins L, Hamarneh W, Ameerally P, Anthony K, Machado L. Duchenne muscular dystrophy gene product expression is associated with survival in head and neck squamous cell carcinoma. Sci Rep 2025; 15:10754. [PMID: 40155657 PMCID: PMC11953289 DOI: 10.1038/s41598-025-94221-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
Mutation of the Duchenne muscular dystrophy (DMD) gene causes neuromuscular disorders, but increasing evidence has implicated DMD in the development and progression of several major cancer types. This study investigates the prognostic and biological significance of DMD expression in head and neck squamous cell carcinoma (HNSCC). Analysis of The Cancer Genome Atlas (TCGA) data revealed that high DMD expression correlates with improved overall (median survival difference: 22 months, p = 0.0083) and progression-free (p = 0.0237) survival. The Dp71ab transcript is most strongly associated with better outcomes (median overall survival: 42 months, p = 0.0007). Notably, DMD expression levels stratify HPV-positive patients, identifying a DMD low/HPV-positive subgroup with poor outcomes. Immunohistochemical analysis of 50 HNSCC tissue cases confirmed dystrophin localisation in the nucleus and cytoplasm, with high nuclear expression linked to longer overall survival (mean difference: 31 months, p = 0.0497). Functional assays in HNSCC cells showed that Dp71ab overexpression disrupts nuclear morphology and reduces proliferation. Differential gene expression analysis additionally identified 388 upregulated and 30 downregulated genes, with pathways linked to muscle processes, ribosome biogenesis and non-coding RNA regulation. These findings highlight DMD as a potential biomarker and/or therapeutic target in HNSCC, warranting further mechanistic studies of Dp71 isoforms.
Collapse
Affiliation(s)
- Leanne Jones
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
- Department of Cancer and Genomic Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Sonika Divakar
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
| | - Lewis Collins
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK
| | - Wael Hamarneh
- Northampton General Hospital NHS Trust, Northampton, NN1 5BD, UK
| | - Phillip Ameerally
- Maxillofacial Department, Northampton General Hospital NHS Trust, Northampton, NN1 5BD, UK
| | - Karen Anthony
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK.
| | - Lee Machado
- Centre for Physical Activity and Life Sciences, University of Northampton, University Drive, Northampton, NN1 5PH, UK.
| |
Collapse
|
7
|
Henzi BC, Baumann D, Michalopoulou E, Erni SJ, Steiner L, Lötscher N, Tscherter A, Klein A. Education and participation in children and adolescents with Duchenne muscular dystrophy in Switzerland. Eur J Paediatr Neurol 2025; 56:107-114. [PMID: 40349652 DOI: 10.1016/j.ejpn.2025.03.010] [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] [Received: 10/15/2024] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 05/14/2025]
Abstract
Quality of life in Duchenne muscular dystrophy has been reported to be negatively affected by the lack of qualifying education and the lack of opportunities for participation in leisure activities. Two thirds of patients with Duchenne muscular dystrophy have cognitive and/or psychiatric problems. Thus, we conducted a survey study on mobility, school problems, executive functions, social participation and quality of life in young patients in Switzerland. We contacted 60 male patients with Duchenne muscular dystrophy aged 8-18 years through the Swiss Registry for Neuromuscular Disorders. Mobility, school problems and social participation in leisure activities were assessed with a self-constructed questionnaire. Quality of life and executive function were assessed using KIDSCREEN-10 and BRIEF scores, respectively. Out of 60 dispatched surveys, 67 % were filled out and included. Approximately half of the participants went to a special needs school, and 83 % rated their overall quality of life as good. We did not find a correlation between mobility and quality of life, whereas more social participation was correlated with higher quality of life. Furthermore, patients with more difficulties in executive functions showed less participation and lower quality of life. These results underline the need for neuropsychological and adapted assistance in patients with Duchenne muscular dystrophy to facilitate education and social participation.
Collapse
Affiliation(s)
- Bettina C Henzi
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland.
| | - Dominique Baumann
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | | | - Sarah J Erni
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Leonie Steiner
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nadine Lötscher
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Anne Tscherter
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Andrea Klein
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Pediatric Neurology and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| |
Collapse
|
8
|
Costa CIS, Madanelo L, Wang JYT, da Silva Campos G, De Sanctis Girardi AC, Scliar M, Monfardini F, de Cássia Mingroni Pavanello R, Cória VR, Vibranovski MD, Krepischi AC, Lourenço NCV, Zatz M, Yamamoto GL, Zachi EC, Passos-Bueno MR. Understanding rare variant contributions to autism: lessons from dystrophin-deficient model. NPJ Genom Med 2025; 10:18. [PMID: 40050609 PMCID: PMC11885547 DOI: 10.1038/s41525-025-00469-5] [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: 02/19/2024] [Accepted: 01/28/2025] [Indexed: 03/09/2025] Open
Abstract
Duchenne and Becker Muscular Dystrophy are dystrophinopathies with a prevalence of 1:5000-6000 males, caused by pathogenic variants in DMD. These conditions are often accompanied by neurodevelopmental disorders (NDDs) like autism (ASD; ~20%) and intellectual disability (ID; ~30%). However, their low penetrance in dystrophinopathies suggests additional contributing factors. In our study, 83 individuals with dystrophinopathies were clinically evaluated and categorized based on ASD (36 individuals), ID risk (12 individuals), or controls (35 individuals). Exome sequencing analysis revealed an enrichment of risk de novo variants (DNVs) in ASD-DMD individuals (adjusted p value = 0.0356), with the number of DNVs correlating with paternal age (p value = 0.0133). Additionally, DMD-ASD individuals showed a higher average of rare risk variants (RRVs) compared to DMD-Controls (adjusted p value = 0.0285). Gene ontology analysis revealed an enrichment of extracellular matrix-related genes, especially collagens, and Ehlers-Danlos syndrome genes in ASD-DMD and DMD-ID groups. These findings support an oligogenic model for ASD in dystrophinopathies, highlighting the importance of investigating homogenized samples to elucidate ASD's genetic architecture.
Collapse
Affiliation(s)
- Claudia Ismania Samogy Costa
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Luciana Madanelo
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Jaqueline Yu Ting Wang
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Gabriele da Silva Campos
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina De Sanctis Girardi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Marília Scliar
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Frederico Monfardini
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Rita de Cássia Mingroni Pavanello
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Vivian Romanholi Cória
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Dulcetti Vibranovski
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Ana Cristina Krepischi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Naila Cristina Vilaça Lourenço
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Mayana Zatz
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Guilherme Lopes Yamamoto
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Elaine Cristina Zachi
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Maria Rita Passos-Bueno
- Departamento de Genética e Biologia Evolutiva, Centro de Estudos do Genoma Humano e Células-tronco, Instituto de Biociências, Universidade de São Paulo, São Paulo, SP, Brasil.
| |
Collapse
|
9
|
Vaillend C, Aoki Y, Mercuri E, Hendriksen J, Tetorou K, Goyenvalle A, Muntoni F. Duchenne muscular dystrophy: recent insights in brain related comorbidities. Nat Commun 2025; 16:1298. [PMID: 39900900 PMCID: PMC11790952 DOI: 10.1038/s41467-025-56644-w] [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: 06/27/2024] [Accepted: 01/20/2025] [Indexed: 02/05/2025] Open
Abstract
Duchenne muscular dystrophy (DMD), the most common childhood muscular dystrophy, arises from DMD gene mutations, affecting the production of muscle dystrophin protein. Brain dystrophin-gene products are also transcribed via internal promoters. Their deficiency contributes to comorbidities, including intellectual disability ( ~ 22% of patients), autism ( ~ 6%) and attention deficit disorders ( ~ 18%), representing a major unmet need for patients and families. Thus, improvement of their diagnosis and treatment is needed. Dystrophic mouse models exhibit similar phenotypes, where genetic therapies restoring brain dystrophins improve their behaviour. This suggests that future genetic therapies could address both muscle and brain dysfunction in DMD patients.
Collapse
Affiliation(s)
- Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400, Saclay, Paris, France
| | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, 187-8502, Japan
| | - Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Jos Hendriksen
- Kempenhaeghe Centre for Neurological Learning Disabilities, Heeze, the Netherlands; Maastricht University, School for Mental Health and Neuroscience, Maastricht, the Netherlands.
| | - Konstantina Tetorou
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Aurelie Goyenvalle
- Université Paris-Saclay, UVSQ, Inserm, END-ICAP, 78000, Versailles, France.
| | - Francesco Muntoni
- University College London Great Ormond Street Institute of Child Health, London, UK.
| |
Collapse
|
10
|
Liu V, Hanson E, Owens JW, Hopkin RJ, Shillington A. A Dual Diagnosis of Okur-Chung Neurodevelopmental Syndrome and Becker Muscular Dystrophy: Inquiry Into the Lower Limits of Neurodevelopmental Functioning Attributable to Muscular Dystrophy. Brain Behav 2025; 15:e70276. [PMID: 39915227 PMCID: PMC11802274 DOI: 10.1002/brb3.70276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 02/11/2025] Open
Abstract
PURPOSE This case discusses the limits of neurodevelopmental functioning attributable to Duchenne's Muscular Dystrophy (DMD) dysfunction. METHOD A 3-year-old male presented with global developmental delay, growth failure, and dysmorphic facial features. An SNP microarray revealed an interstitial duplication in exon 55 of DMD suggestive of Becker Muscular Dystrophy (BMD), but his degree of delays led to follow-up exome sequencing revealing a pathogenic CSNK2A1 variant diagnostic for Okur-Chung Neurodevelopmental Syndrome. FINDINGS Large cohorts predict a full-scale IQ (FSIQ) of 88.3 ± 13.9 among all patients with BMD and 86.1 ± 15.0 among all patients with DMD, while variants impacting the brain dystrophin isoform Dp140 are associated with FSIQ of 77.7 ± 10.8 in BMD and 78.8 ± 18.6 in DMD. CONCLUSION An FSIQ one standard deviation below these expected ranges should prompt screening for alternative causes of neurodevelopmental delays, and an FSIQ two standard deviations below these ranges should prompt broad-spectrum genetic testing.
Collapse
Affiliation(s)
- Victoria Liu
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Eva Hanson
- University of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Joshua W Owens
- Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Robert J. Hopkin
- Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Amelle Shillington
- Division of Human GeneticsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| |
Collapse
|
11
|
Konieczny P. Systemic Treatment of Body-Wide Duchenne Muscular Dystrophy Symptoms. Clin Pharmacol Ther 2024; 116:1472-1484. [PMID: 38965715 DOI: 10.1002/cpt.3363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
12
|
González-Reyes M, Aragón J, Sánchez-Trujillo A, Rodríguez-Martínez G, Duarte K, Eleftheriou E, Barnier JV, Naquin D, Thermes C, Romo-Yáñez J, Roger JE, Rendon A, Vaillend C, Montanez C. Expression of Dystrophin Dp71 Splice Variants Is Temporally Regulated During Rodent Brain Development. Mol Neurobiol 2024; 61:10883-10900. [PMID: 38802640 PMCID: PMC11584426 DOI: 10.1007/s12035-024-04232-2] [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: 12/12/2023] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Abstract
Dystrophin Dp71 is the major product of the Duchenne muscular dystrophy (DMD) gene in the brain, and its loss in DMD patients and mouse models leads to cognitive impairments. Dp71 is expressed as a range of proteins generated by alternative splicing of exons 71 to 74 and 78, classified in the main Dp71d and Dp71f groups that contain specific C-terminal ends. However, it is unknown whether each isoform has a specific role in distinct cell types, brain regions, and/or stages of brain development. In the present study, we characterized the expression of Dp71 isoforms during fetal (E10.5, E15.5) and postnatal (P1, P7, P14, P21 and P60) mouse and rat brain development. We finely quantified the expression of several Dp71 transcripts by RT-PCR and cloning assays in samples from whole-brain and distinct brain structures. The following Dp71 transcripts were detected: Dp71d, Dp71d∆71, Dp71d∆74, Dp71d∆71,74, Dp71d∆71-74, Dp71f, Dp71f∆71, Dp71f∆74, Dp71f∆71,74, and Dp71fΔ71-74. We found that the Dp71f isoform is the main transcript expressed at E10.5 (> 80%), while its expression is then progressively reduced and replaced by the expression of isoforms of the Dp71d group from E15.5 to postnatal and adult ages. This major finding was confirmed by third-generation nanopore sequencing. In addition, we found that the level of expression of specific Dp71 isoforms varies as a function of postnatal stages and brain structure. Our results suggest that Dp71 isoforms have different and complementary roles during embryonic and postnatal brain development, likely taking part in a variety of maturation processes in distinct cell types.
Collapse
Affiliation(s)
- Mayram González-Reyes
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, Saclay, 91400, France
| | - Jorge Aragón
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
- Institut de la Vision, Sorbonne Université-INSERM-CNRS, 17 rue Moreau, Paris, 75012, France
| | - Alejandra Sánchez-Trujillo
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
| | - Griselda Rodríguez-Martínez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| | - Kevin Duarte
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, Saclay, 91400, France
| | - Evangelia Eleftheriou
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, 91198, France
| | - Jean-Vianney Barnier
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, Saclay, 91400, France
| | - Delphine Naquin
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, 91198, France
| | - Claude Thermes
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, 91198, France
| | - José Romo-Yáñez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico
- Institut de la Vision, Sorbonne Université-INSERM-CNRS, 17 rue Moreau, Paris, 75012, France
- Coordinación de Endocrinología Ginecológica y Perinatal, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - Jérome E Roger
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, Saclay, 91400, France
- CERTO-Retina France, Saclay, 91400, France
| | - Alvaro Rendon
- Institut de la Vision, Sorbonne Université-INSERM-CNRS, 17 rue Moreau, Paris, 75012, France
| | - Cyrille Vaillend
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, Saclay, 91400, France.
| | - Cecilia Montanez
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Mexico City, Mexico.
| |
Collapse
|
13
|
Neuhoff K, Kilicarslan OA, Preuße C, Zaum AK, Kölbel H, Lochmüller H, Schara-Schmidt U, Polavarapu K, Roos A, Gangfuß A. Expanding the Molecular Genetic Landscape of Dystrophinopathies and Associated Phenotypes. Biomedicines 2024; 12:2738. [PMID: 39767645 PMCID: PMC11727156 DOI: 10.3390/biomedicines12122738] [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: 10/09/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: X-linked dystrophinopathies are a group of neuromuscular diseases caused by pathogenic variants in the DMD gene (MIM *300377). Duchenne muscular dystrophy (DMD; MIM #310200) is the most common inherited muscular dystrophy. Methods: We screened datasets of 403 male, genetically confirmed X-linked dystrophinopathy patients and identified 13 pathogenic variants of the DMD gene that have not been described in the literature thus far. For all patients we provide additional data on the clinical course, genotype-phenotype correlations as well as histological datasets of nine patients. In two cases, we used RNA-Seq analyses, showing that this method can be particularly helpful in cases of deep intrinsic variants. Results: We were able to show, that a combination of the different datasets is helpful to counsel families and provides a better understanding of the underlying pathophysiology. Conclusions: Overall, we elaborated upon the persistent challenge of determining the course of disease from genetic analysis alone, rather supporting the concept of a clinical continuum of dystrophinopathies with our combined clinical, histological and molecular genetic findings.
Collapse
Affiliation(s)
- Katja Neuhoff
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (K.N.); (H.K.); (U.S.-S.); (A.R.)
| | - Ozge Aksel Kilicarslan
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 5B2, Canada; (O.A.K.); (H.L.)
| | - Corinna Preuße
- Department of Neuropathology, Charité-University Medicine Berlin, 10117 Berlin, Germany;
| | - Ann-Kathrin Zaum
- Institute of Human Genetics, University of Würzburg, 97074 Würzburg, Germany;
| | - Heike Kölbel
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (K.N.); (H.K.); (U.S.-S.); (A.R.)
| | - Hanns Lochmüller
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 5B2, Canada; (O.A.K.); (H.L.)
- Centro Nacional de Análisis Genómico (CNAG-CRG), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- Brain and Mind Research Institute, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON K1H 8L6, Canada
- Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, Medical Center, University of Freiburg, 79106 Freiburg, Germany
| | - Ulrike Schara-Schmidt
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (K.N.); (H.K.); (U.S.-S.); (A.R.)
| | - Kiran Polavarapu
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 5B2, Canada; (O.A.K.); (H.L.)
| | - Andreas Roos
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (K.N.); (H.K.); (U.S.-S.); (A.R.)
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 5B2, Canada; (O.A.K.); (H.L.)
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, 40225 Duesseldorf, Germany
| | - Andrea Gangfuß
- Department of Pediatric Neurology, Centre for Neuromuscular Disorders, Centre for Translational Neuro- and Behavioral Sciences, University Duisburg-Essen, 45122 Essen, Germany; (K.N.); (H.K.); (U.S.-S.); (A.R.)
| |
Collapse
|
14
|
Prigogine C, Ruiz JM, Cebolla AM, Deconinck N, Servais L, Gailly P, Dan B, Cheron G. Cerebellar dysfunction in the mdx mouse model of Duchenne muscular dystrophy: An electrophysiological and behavioural study. Eur J Neurosci 2024; 60:6470-6489. [PMID: 39415418 DOI: 10.1111/ejn.16566] [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: 03/12/2024] [Revised: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 10/18/2024]
Abstract
Patients with Duchenne muscular dystrophy (DMD) commonly show specific cognitive deficits in addition to a severe muscle impairment caused by the absence of dystrophin expression in skeletal muscle. These cognitive deficits have been related to the absence of dystrophin in specific regions of the central nervous system, notably cerebellar Purkinje cells (PCs). Dystrophin has recently been involved in GABAA receptors clustering at postsynaptic densities, and its absence, by disrupting this clustering, leads to decreased inhibitory input to PC. We performed an in vivo electrophysiological study of the dystrophin-deficient muscular dystrophy X-linked (mdx) mouse model of DMD to compare PC firing and local field potential (LFP) in alert mdx and control C57Bl/10 mice. We found that the absence of dystrophin is associated with altered PC firing and the emergence of fast (~160-200 Hz) LFP oscillations in the cerebellar cortex of alert mdx mice. These abnormalities were not related to the disrupted expression of calcium-binding proteins in cerebellar PC. We also demonstrate that cerebellar long-term depression is altered in alert mdx mice. Finally, mdx mice displayed a force weakness, mild impairment of motor coordination and balance during behavioural tests. These findings demonstrate the existence of cerebellar dysfunction in mdx mice. A similar cerebellar dysfunction may contribute to the cognitive deficits observed in patients with DMD.
Collapse
Affiliation(s)
- Cynthia Prigogine
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Electrophysiology, Université de Mons, Mons, Belgium
| | | | - Ana Maria Cebolla
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium
| | | | - Philippe Gailly
- Laboratory of Cell Physiology, Université Catholique de Louvain, Brussels, Belgium
| | - Bernard Dan
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Rehabilitation Hospital Inkendaal, Vlezenbeek, Belgium
| | - Guy Cheron
- Laboratory of Neurophysiology and Movement Biomechanics, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Electrophysiology, Université de Mons, Mons, Belgium
| |
Collapse
|
15
|
Fujimoto T, Okamura T, Itoh K. Extraction method combining saponin and trehalose useful for analyzing fragile intermolecular association. Biochem Biophys Res Commun 2024; 727:150323. [PMID: 38945065 DOI: 10.1016/j.bbrc.2024.150323] [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: 06/24/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Immunoprecipitation (IP) and co-immunoprecipitation (co-IP) are well-established methodologies to analyze protein expression and intermolecular interaction. Composition of extraction and washing buffer for preparing protein is important to accomplish experimental purpose. Various kinds of detergents are included in buffer to adjust extraction efficiency and washing effect. Among them, Triton X-100 (Tx-100), Nonidet P-40 (NP40), deoxycholic acid (DOC) and SDS are generally used according to experimental purpose and characteristic features of protein of interest. In some cases, general detergents disrupt intermolecular interaction and make it impossible to analyze molecular relation of protein of interest with its binding partners. In this study, we propose saponin, a natural detergent, is useful for co-immunoprecipitation when analyzing fragile intermolecular interactions, in which dystrophin and dystroglycan are used as a representative interaction. One of the most notable findings in this report is that intermolecular association between dystrophin and dystroglycan is maintained in saponin buffer whereas general detergents, such as Tx-100, NP40 and DOC, dissociate its binding. Furthermore, supplementation of trehalose, which has been shown to act as a molecular chaperone, facilitates efficient detection of dystrophin-dystroglycan macromolecular complex in co-IP assay. Importantly, the extraction buffer comprising 3 % saponin, 0.5 M trehalose and 0.05 % Tx-100 (we named it STX buffer) is applicable to co-IP for another molecular interaction, N-cadherin and β-catenin, indicating that this methodology can be used for versatile proteins of interest. Thus, STX buffer emerges as an alternative extraction method useful for analyzing fragile intermolecular associations and provides opportunity to identify complex interactomes, which may facilitate proteome-research and functional analysis of proteins of interest.
Collapse
Affiliation(s)
- Takahiro Fujimoto
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine (NCGM), Tokyo, 162-8655, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
16
|
Yang J, Cao C, Liu J, Liu Y, Lu J, Yu H, Li X, Wu J, Yu Z, Li H, Chen G. Dystrophin 71 deficiency causes impaired aquaporin-4 polarization contributing to glymphatic dysfunction and brain edema in cerebral ischemia. Neurobiol Dis 2024; 199:106586. [PMID: 38950712 DOI: 10.1016/j.nbd.2024.106586] [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: 03/27/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/03/2024] Open
Abstract
OBJECTIVE The glymphatic system serves as a perivascular pathway that aids in clearing liquid and solute waste from the brain, thereby enhancing neurological function. Disorders in glymphatic drainage contribute to the development of vasogenic edema following cerebral ischemia, although the molecular mechanisms involved remain poorly understood. This study aims to determine whether a deficiency in dystrophin 71 (DP71) leads to aquaporin-4 (AQP4) depolarization, contributing to glymphatic dysfunction in cerebral ischemia and resulting in brain edema. METHODS A mice model of middle cerebral artery occlusion and reperfusion was used. A fluorescence tracer was injected into the cortex and evaluated glymphatic clearance. To investigate the role of DP71 in maintaining AQP4 polarization, an adeno-associated virus with the astrocyte promoter was used to overexpress Dp71. The expression and distribution of DP71 and AQP4 were analyzed using immunoblotting, immunofluorescence, and co-immunoprecipitation techniques. The behavior ability of mice was evaluated by open field test. Open-access transcriptome sequencing data were used to analyze the functional changes of astrocytes after cerebral ischemia. MG132 was used to inhibit the ubiquitin-proteasome system. The ubiquitination of DP71 was detected by immunoblotting and co-immunoprecipitation. RESULTS During the vasogenic edema stage following cerebral ischemia, a decline in the efflux of interstitial fluid tracer was observed. DP71 and AQP4 were co-localized and interacted with each other in the perivascular astrocyte endfeet. After cerebral ischemia, there was a notable reduction in DP71 protein expression, accompanied by AQP4 depolarization and proliferation of reactive astrocytes. Increased DP71 expression restored glymphatic drainage and reduced brain edema. AQP4 depolarization, reactive astrocyte proliferation, and the behavior of mice were improved. After cerebral ischemia, DP71 was degraded by ubiquitination, and MG132 inhibited the decrease of DP71 protein level. CONCLUSION AQP4 depolarization after cerebral ischemia leads to glymphatic clearance disorder and aggravates cerebral edema. DP71 plays a pivotal role in regulating AQP4 polarization and consequently influences glymphatic function. Changes in DP71 expression are associated with the ubiquitin-proteasome system. This study offers a novel perspective on the pathogenesis of brain edema following cerebral ischemia.
Collapse
Affiliation(s)
- Jian Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiale Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Yangyang Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - HaoYun Yu
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China.
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China; Institute of Stroke Research, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
17
|
Chang IYF, Tsai HC, Chen CH, Chen HC, Huang CW, Cox GF, Huang FM, Lin YY, Chen KT, Lin YJ, Wei KC. CAN008 prolongs overall survival in patients with newly diagnosed GBM characterized by high tumor mutational burden. Biomed J 2024; 47:100660. [PMID: 37741340 PMCID: PMC11340566 DOI: 10.1016/j.bj.2023.100660] [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: 05/29/2023] [Revised: 07/31/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND A previous phase 1 dose-escalation study in Taiwan indicated CAN008 (asunercept) with standard concurrent chemoradiotherapy (CCRT) improved progression-free survival (PFS) in newly diagnosed glioblastoma (GBM) patients. This study evaluates the efficacy of CAN008 in promoting overall survival (OS) and identifies genetic alterations associated with treatment responses. METHODS We compared OS of 5-year follow-ups from 9 evaluable CAN008 cohort patients (6 received high-dose and 3 received low-dose) to a historical Taiwanese GBM cohort with 164 newly diagnosed patients. CAN008 treatment response-associated genetic alterations were identified by whole-exome sequencing and comparing variant differences between response groups. Associations among patient survival, tumor mutational burden (TMB), and genetic alterations were analyzed using CAN008 cohort and TCGA-GBM dataset. RESULTS OS for high-dose CAN008 patients at 2 and 5 years was 83% and 67%, respectively, and 40.1% and 8.8% for the historical GBM cohort, respectively. Better OS was observed in the high-dose CAN008 cohort (without reaching the median survival) than the historical GBM cohort (median OS: 20 months; p = 0.0103). Five high-dose CAN008 patients were divided into good and poor response groups based on their PFS. A higher variant count and TMB were observed in good response patients, whereas no significant association was observed between TMB and patient survival in the newly diagnosed TCGA-GBM dataset, suggesting TMB may modulate patient CAN008 response. CONCLUSION CAN008 combined with standard CCRT treatment prolonged the PFS and OS of newly diagnosed GBM patients compared to standard therapy alone. Higher treatment efficacy was associated with higher TMB.
Collapse
Affiliation(s)
- Ian Yi-Feng Chang
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Hong-Chieh Tsai
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Chia-Hua Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu-Chi Chen
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Wen Huang
- School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | | | | | - You-Yu Lin
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei, Taiwan
| | - Ko-Ting Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Ya-Jui Lin
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
| | - Kuo-Chen Wei
- School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan; Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan.
| |
Collapse
|
18
|
Wu L, Zheng H, Guo X, Li N, Qin L, Li X, Lou G. Integrative analyses of genes associated with oxidative stress and cellular senescence in triple-negative breast cancer. Heliyon 2024; 10:e34524. [PMID: 39130410 PMCID: PMC11315143 DOI: 10.1016/j.heliyon.2024.e34524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 07/06/2024] [Accepted: 07/10/2024] [Indexed: 08/13/2024] Open
Abstract
Background Oxidative stress and cellular senescence (OSCS) have great impacts on the occurrence and progression of triple-negative breast cancer (TNBC). This study was intended to construct a prognostic model based on oxidative stress and cellular senescence related difference expression genes (OSCSRDEGs) for TNBC. Methods The Cancer Genome Atlas (TCGA) databases and two Gene Expression Omnibus (GEO) databases were used to identify OSCSRDEGs. The relationship between OSCSRDEGs and immune infiltration was examined using single-sample gene-set enrichment analysis (ssGSEA), ESTIMATE, and the CIBERSORT algorithm. Least absolute shrinkage and selection operator (LASSO) regression analyses, Cox regression and Kaplan-Meier analysis were employed to construct a prognostic model. Receiver operating characteristic (ROC) curves, nomograms, and decision curve analysis (DCA) were used to evaluate the prognostic efficacy. Gene Set Enrichment Analysis (GSEA) Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) were utilized to explore the potential functions and mechanism. Results A comprehensive analysis identified a total of 27 OSCSRDEGs, out of which 15 genes selected for development of a prognostic model. A high degree of statistical significance was observed for the riskscores derived from this model to accurately predict TNBC Overall survival. The decision curve analysis (DCA) and ROC curve analysis further confirmed the superior accuracy of the OSCSRDEGs prognostic model in predicting efficacy. Notably, the nomogram analysis highlighted that DMD exhibited the highest utility within the model. In comparison between high and low OSCScore groups, the infiltration abundance of immune cells was statistically different in the TCGA-TNBC dataset. Conclusion These studies have effectively identified four essential OSCSRDEGs (CFI, DMD, NDRG2, and NRP1) and meticulously developed an OSCS-associated prognostic model for individuals diagnosed with TNBC. These discoveries have the potential to significantly contribute to the comprehension of the involvement of OSCS in TNBC.
Collapse
Affiliation(s)
- Lihua Wu
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Hongyan Zheng
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xiaorong Guo
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Nan Li
- Department of Pathology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Luyao Qin
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xiaoqing Li
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Ge Lou
- Department of Pathology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| |
Collapse
|
19
|
Alnassar N, Hajto J, Rumney RMH, Verma S, Borczyk M, Saha C, Kanczler J, Butt AM, Occhipinti A, Pomeroy J, Angione C, Korostynski M, Górecki DC. Ablation of the dystrophin Dp71f alternative C-terminal variant increases sarcoma tumour cell aggressiveness. Hum Mol Genet 2024:ddae094. [PMID: 38850567 DOI: 10.1093/hmg/ddae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 05/08/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024] Open
Abstract
Alterations in Dp71 expression, the most ubiquitous dystrophin isoform, have been associated with patient survival across tumours. Intriguingly, in certain malignancies, Dp71 acts as a tumour suppressor, while manifesting oncogenic properties in others. This diversity could be explained by the expression of two Dp71 splice variants encoding proteins with distinct C-termini, each with specific properties. Expression of these variants has impeded the exploration of their unique roles. Using CRISPR/Cas9, we ablated the Dp71f variant with the alternative C-terminus in a sarcoma cell line not expressing the canonical C-terminal variant, and conducted molecular (RNAseq) and functional characterisation of the knockout cells. Dp71f ablation induced major transcriptomic alterations, particularly affecting the expression of genes involved in calcium signalling and ECM-receptor interaction pathways. The genome-scale metabolic analysis identified significant downregulation of glucose transport via membrane vesicle reaction (GLCter) and downregulated glycolysis/gluconeogenesis pathway. Functionally, these molecular changes corresponded with, increased calcium responses, cell adhesion, proliferation, survival under serum starvation and chemotherapeutic resistance. Knockout cells showed reduced GLUT1 protein expression, survival without attachment and their migration and invasion in vitro and in vivo were unaltered, despite increased matrix metalloproteinases release. Our findings emphasise the importance of alternative splicing of dystrophin transcripts and underscore the role of the Dp71f variant, which appears to govern distinct cellular processes frequently dysregulated in tumour cells. The loss of this regulatory mechanism promotes sarcoma cell survival and treatment resistance. Thus, Dp71f is a target for future investigations exploring the intricate functions of specific DMD transcripts in physiology and across malignancies.
Collapse
Affiliation(s)
- Nancy Alnassar
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Jacek Hajto
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Chandrika Saha
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Janos Kanczler
- Bone & Joint Research Group, Department of Human Development and Health, University of Southampton, Tremona Road, Southampton SO16 6YD, United Kingdom
| | - Arthur M Butt
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Joanna Pomeroy
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside University, Middlesbrough, Tees Valley TS1 3BX, United Kingdom
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PAS, Smetna 12, Krakow 31155, Poland
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth PO1 2DT, United Kingdom
| |
Collapse
|
20
|
Wang Y, Wen X, Shen XM, Di L, Sun Y, Li Y, Zhang S, Wen Q, Wang J, Duo J, Huang Y, Lu Y, Xu M, Wang M, Chen H, Zhu W, Da Y. A rare complex structural variant of novel intragenic inversion combined with reciprocal translocation t(X;1)(p21.2;p13.3) in Duchenne muscular dystrophy. Neuromuscul Disord 2024; 39:24-29. [PMID: 38714145 DOI: 10.1016/j.nmd.2024.04.003] [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: 01/18/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 05/09/2024]
Abstract
Structural variants (SVs) are infrequently observed in Duchenne muscular dystrophy (DMD), a condition mainly marked by deletions and point mutations in the DMD gene. SVs in DMD remain difficult to reliably detect due to the limited SV-detection capacity of conventionally used short-read sequencing technology. Herein, we present a family, a boy and his mother, with clinical signs of muscular dystrophy, elevated creatinine kinase levels, and intellectual disability. A muscle biopsy from the boy showed dystrophin deficiency. Routine molecular techniques failed to detect abnormalities in the DMD gene, however, dystrophin mRNA transcripts analysis revealed an absence of exons 59 to 79. Subsequent long-read whole-genome sequencing identified a rare complex structural variant, a 77 kb novel intragenic inversion, and a balanced translocation t(X;1)(p21.2;p13.3) rearrangement within the DMD gene, expanding the genetic spectrum of dystrophinopathy. Our findings suggested that SVs should be considered in cases where conventional molecular techniques fail to identify pathogenic variants.
Collapse
Affiliation(s)
- Yaye Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Xinmei Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Xin-Ming Shen
- Department of Neurology and Neuromuscular Research Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | - Li Di
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Yanan Sun
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Yun Li
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Shu Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Qi Wen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Jingsi Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Jianying Duo
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Yue Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Yan Lu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Min Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Min Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Hai Chen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Wenjia Zhu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China
| | - Yuwei Da
- Department of Neurology, Xuanwu Hospital, Capital Medical University, No. 45 Changchun Street, Xicheng District, Beijing 100053, China.
| |
Collapse
|
21
|
Vacca O, Zarrouki F, Izabelle C, Belmaati Cherkaoui M, Rendon A, Dalkara D, Vaillend C. AAV-Mediated Restoration of Dystrophin-Dp71 in the Brain of Dp71-Null Mice: Molecular, Cellular and Behavioral Outcomes. Cells 2024; 13:718. [PMID: 38667332 PMCID: PMC11049308 DOI: 10.3390/cells13080718] [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: 02/29/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
A deficiency in the shortest dystrophin-gene product, Dp71, is a pivotal aggravating factor for intellectual disabilities in Duchenne muscular dystrophy (DMD). Recent advances in preclinical research have achieved some success in compensating both muscle and brain dysfunctions associated with DMD, notably using exon skipping strategies. However, this has not been studied for distal mutations in the DMD gene leading to Dp71 loss. In this study, we aimed to restore brain Dp71 expression in the Dp71-null transgenic mouse using an adeno-associated virus (AAV) administrated either by intracardiac injections at P4 (ICP4) or by bilateral intracerebroventricular (ICV) injections in adults. ICP4 delivery of the AAV9-Dp71 vector enabled the expression of 2 to 14% of brain Dp71, while ICV delivery enabled the overexpression of Dp71 in the hippocampus and cortex of adult mice, with anecdotal expression in the cerebellum. The restoration of Dp71 was mostly located in the glial endfeet that surround capillaries, and it was associated with partial localization of Dp71-associated proteins, α1-syntrophin and AQP4 water channels, suggesting proper restoration of a scaffold of proteins involved in blood-brain barrier function and water homeostasis. However, this did not result in significant improvements in behavioral disturbances displayed by Dp71-null mice. The potential and limitations of this AAV-mediated strategy are discussed. This proof-of-concept study identifies key molecular markers to estimate the efficiencies of Dp71 rescue strategies and opens new avenues for enhancing gene therapy targeting cognitive disorders associated with a subgroup of severely affected DMD patients.
Collapse
Affiliation(s)
- Ophélie Vacca
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Faouzi Zarrouki
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Charlotte Izabelle
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Mehdi Belmaati Cherkaoui
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| | - Alvaro Rendon
- Department of Therapeutics, Sorbonne University, Institut de la Vision, 75012 Paris, France; (A.R.)
| | - Deniz Dalkara
- Department of Therapeutics, Sorbonne University, Institut de la Vision, 75012 Paris, France; (A.R.)
| | - Cyrille Vaillend
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, 91400 Saclay, France (M.B.C.)
| |
Collapse
|
22
|
Da Cunha D, Miro J, Van Goethem C, Notarnicola C, Hugon G, Carnac G, Cossée M, Koenig M, Tuffery-Giraud S. The exon junction complex is required for DMD gene splicing fidelity and myogenic differentiation. Cell Mol Life Sci 2024; 81:150. [PMID: 38512499 PMCID: PMC10957711 DOI: 10.1007/s00018-024-05188-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Deposition of the exon junction complex (EJC) upstream of exon-exon junctions helps maintain transcriptome integrity by preventing spurious re-splicing events in already spliced mRNAs. Here we investigate the importance of EJC for the correct splicing of the 2.2-megabase-long human DMD pre-mRNA, which encodes dystrophin, an essential protein involved in cytoskeletal organization and cell signaling. Using targeted RNA-seq, we show that knock-down of the eIF4A3 and Y14 core components of EJC in a human muscle cell line causes an accumulation of mis-splicing events clustered towards the 3' end of the DMD transcript (Dp427m). This deregulation is conserved in the short Dp71 isoform expressed ubiquitously except in adult skeletal muscle and is rescued with wild-type eIF4A3 and Y14 proteins but not with an EJC assembly-defective mutant eIF4A3. MLN51 protein and EJC-associated ASAP/PSAP complexes independently modulate the inclusion of the regulated exons 71 and 78. Our data confirm the protective role of EJC in maintaining splicing fidelity, which in the DMD gene is necessary to preserve the function of the critical C-terminal protein-protein interaction domain of dystrophin present in all tissue-specific isoforms. Given the role of the EJC in maintaining the integrity of dystrophin, we asked whether the EJC could also be involved in the regulation of a mechanism as complex as skeletal muscle differentiation. We found that eIF4A3 knockdown impairs myogenic differentiation by blocking myotube formation. Collectively, our data provide new insights into the functional roles of EJC in human skeletal muscle.
Collapse
Affiliation(s)
- Dylan Da Cunha
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Julie Miro
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Charles Van Goethem
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France
- Montpellier BioInformatique Pour Le Diagnostic Clinique (MOBIDIC), Plateau de Médecine Moléculaire Et Génomique (PMMG), CHU Montpellier, 34295, Montpellier, France
| | | | - Gérald Hugon
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Gilles Carnac
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Mireille Cossée
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France
| | - Michel Koenig
- PhyMedExp, Univ Montpellier, CNRS, INSERM, Montpellier, France
- Laboratoire de Génétique Moléculaire, CHU de Montpellier, Montpellier, France
| | | |
Collapse
|
23
|
Simon KC, Cadle C, Nakra N, Nagel MC, Malerba P. Age-associated sleep spindle characteristics in Duchenne muscular dystrophy. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae015. [PMID: 38525359 PMCID: PMC10960605 DOI: 10.1093/sleepadvances/zpae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 12/18/2023] [Indexed: 03/26/2024]
Abstract
Brain oscillations of non-rapid eye movement sleep, including slow oscillations (SO, 0.5-1.5 Hz) and spindles (10-16 Hz), mirror underlying brain maturation across development and are associated with cognition. Hence, age-associated emergence and changes in the electrophysiological properties of these rhythms can lend insight into cortical development, specifically in comparisons between pediatric populations and typically developing peers. We previously evaluated age-associated changes in SOs in male patients with Duchenne muscular dystrophy (DMD), finding a significant age-related decline between 4 and 18 years. While primarily a muscle disorder, male patients with DMD can also have sleep, cognitive, and cortical abnormalities, thought to be driven by altered dystrophin expression in the brain. In this follow-up study, we characterized the age-associated changes in sleep spindles. We found that age-dependent spindle characteristics in patients with DMD, including density, frequency, amplitude, and duration, were consistent with age-associated trends reported in the literature for typically developing controls. Combined with our prior finding of age-associated decline in SOs, our results suggest that SOs, but not spindles, are a candidate intervention target to enhance sleep in patients with DMD.
Collapse
Affiliation(s)
- Katharine C Simon
- Department of Pediatrics, School of Medicine, University of California, Irvine, Irvine, CA, USA
- Pulmonology Department, Children’s Hospital of Orange County, Orange, CA, USA
| | - Chelsea Cadle
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
| | - Neal Nakra
- Pulmonology Department, Children’s Hospital of Orange County, Orange, CA, USA
| | - Marni C Nagel
- Department of Pediatric Psychology, Children’s Hospital of Orange County, Orange, CA, USA
| | - Paola Malerba
- Center for Biobehavioral Health, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics, School of Medicine, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
24
|
Jahncke JN, Miller DS, Krush M, Schnell E, Wright KM. Inhibitory CCK+ basket synapse defects in mouse models of dystroglycanopathy. eLife 2024; 12:RP87965. [PMID: 38179984 PMCID: PMC10942650 DOI: 10.7554/elife.87965] [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] [Indexed: 01/06/2024] Open
Abstract
Dystroglycan (Dag1) is a transmembrane glycoprotein that links the extracellular matrix to the actin cytoskeleton. Mutations in Dag1 or the genes required for its glycosylation result in dystroglycanopathy, a type of congenital muscular dystrophy characterized by a wide range of phenotypes including muscle weakness, brain defects, and cognitive impairment. We investigated interneuron (IN) development, synaptic function, and associated seizure susceptibility in multiple mouse models that reflect the wide phenotypic range of dystroglycanopathy neuropathology. Mice that model severe dystroglycanopathy due to forebrain deletion of Dag1 or Pomt2, which is required for Dystroglycan glycosylation, show significant impairment of CCK+/CB1R+ IN development. CCK+/CB1R+ IN axons failed to properly target the somatodendritic compartment of pyramidal neurons in the hippocampus, resulting in synaptic defects and increased seizure susceptibility. Mice lacking the intracellular domain of Dystroglycan have milder defects in CCK+/CB1R+ IN axon targeting, but exhibit dramatic changes in inhibitory synaptic function, indicating a critical postsynaptic role of this domain. In contrast, CCK+/CB1R+ IN synaptic function and seizure susceptibility was normal in mice that model mild dystroglycanopathy due to partially reduced Dystroglycan glycosylation. Collectively, these data show that inhibitory synaptic defects and elevated seizure susceptibility are hallmarks of severe dystroglycanopathy, and show that Dystroglycan plays an important role in organizing functional inhibitory synapse assembly.
Collapse
Affiliation(s)
- Jennifer N Jahncke
- Neuroscience Graduate Program, Oregon Health & Science UniversityPortlandUnited States
| | - Daniel S Miller
- Neuroscience Graduate Program, Oregon Health & Science UniversityPortlandUnited States
| | - Milana Krush
- Neuroscience Graduate Program, Oregon Health & Science UniversityPortlandUnited States
| | - Eric Schnell
- Operative Care Division, Portland VA Health Care SystemPortlandUnited States
- Anesthesiology and Perioperative Medicine, Oregon Health & Science UniversityPortlandUnited States
| | - Kevin M Wright
- Vollum Institute, Oregon Health & Science UniversityPortlandUnited States
| |
Collapse
|
25
|
García I, Martínez O, López-Paz JF, García M, Espinosa-Blanco P, Rodríguez AA, Pallarès-Sastre M, Ruiz de Lazcano A, Amayra I. Social cognition in DMD and BMD dystrophinopathies: A cross-sectional preliminary study. Clin Neuropsychol 2024; 38:219-234. [PMID: 37081823 DOI: 10.1080/13854046.2023.2202332] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/07/2023] [Indexed: 04/22/2023]
Abstract
Objective: The dystrophinopathies called Duchenne and Becker muscular dystrophies (DMD/BMD) are rare, progressive, incurable, and life-limiting paediatric-onset neuromuscular diseases. These diseases have long been associated with specific neuropsychological deficits. However, the performance of these patients in the social cognition domain has not been properly investigated. Thus, the main objective of this study was to compare the performance on social cognition between DMD/BMD patients and healthy age-matched boys. Method: This cross-sectional study included 20 DMD/BMD children and adolescents and 20 healthy controls. The protocol included the Social Perception Domain of the NEPSY-II, the Reading the Mind in the Eyes Test - Child and Happé's Strange Stories test. General intelligence was controlled to eliminate the possible influence of covariables. All the assessments were performed remotely. Results: Most social cognition tasks were worse in patients with DMD/BMD than in matched healthy controls. These differences remained even after controlling for the general intelligence variable, with the exception of Total Disgust Errors (F = 1.462, p = .234, η2p= .038) and Verbal task (F = 1.820, p = .185, η2p= .047) scores from the NEPSY-II. Conclusions: This is the first study to demonstrate that the neuropsychological domain of social cognition is impaired in DMD/BMD patients, independent of the level of general intelligence. Screening assessments in DMD/BMD patients should be promoted to allow social cognition difficulties to be detected at an early stage to enhance patients' quality of life and social development.
Collapse
Affiliation(s)
- Irune García
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Oscar Martínez
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Juan Francisco López-Paz
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Maitane García
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Patricia Espinosa-Blanco
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Alicia Aurora Rodríguez
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Mercè Pallarès-Sastre
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Aitana Ruiz de Lazcano
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| | - Imanol Amayra
- Neuro-e-Motion Research Team, Department of Psychology, Faculty of Health Sciences, University of Deusto, Bilbao, Spain
| |
Collapse
|
26
|
Szwec S, Kapłucha Z, Chamberlain JS, Konieczny P. Dystrophin- and Utrophin-Based Therapeutic Approaches for Treatment of Duchenne Muscular Dystrophy: A Comparative Review. BioDrugs 2024; 38:95-119. [PMID: 37917377 PMCID: PMC10789850 DOI: 10.1007/s40259-023-00632-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2023] [Indexed: 11/04/2023]
Abstract
Duchenne muscular dystrophy is a devastating disease that leads to progressive muscle loss and premature death. While medical management focuses mostly on symptomatic treatment, decades of research have resulted in first therapeutics able to restore the affected reading frame of dystrophin transcripts or induce synthesis of a truncated dystrophin protein from a vector, with other strategies based on gene therapy and cell signaling in preclinical or clinical development. Nevertheless, recent reports show that potentially therapeutic dystrophins can be immunogenic in patients. This raises the question of whether a dystrophin paralog, utrophin, could be a more suitable therapeutic protein. Here, we compare dystrophin and utrophin amino acid sequences and structures, combining published data with our extended in silico analyses. We then discuss these results in the context of therapeutic approaches for Duchenne muscular dystrophy. Specifically, we focus on strategies based on delivery of micro-dystrophin and micro-utrophin genes with recombinant adeno-associated viral vectors, exon skipping of the mutated dystrophin pre-mRNAs, reading through termination codons with small molecules that mask premature stop codons, dystrophin gene repair by clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9)-mediated genetic engineering, and increasing utrophin levels. Our analyses highlight the importance of various dystrophin and utrophin domains in Duchenne muscular dystrophy treatment, providing insights into designing novel therapeutic compounds with improved efficacy and decreased immunoreactivity. While the necessary actin and β-dystroglycan binding sites are present in both proteins, important functional distinctions can be identified in these domains and some other parts of truncated dystrophins might need redesigning due to their potentially immunogenic qualities. Alternatively, therapies based on utrophins might provide a safer and more effective approach.
Collapse
Affiliation(s)
- Sylwia Szwec
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Zuzanna Kapłucha
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland
| | - Jeffrey S Chamberlain
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, 98109-8055, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Washington School of Medicine, Seattle, WA, 98109-8055, USA
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA, 98109-8055, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98109-8055, USA
| | - Patryk Konieczny
- Institute of Human Biology and Evolution, Faculty of Biology, Adam Mickiewicz University, ul. Uniwersytetu Poznańskiego 6, 61-614, Poznań, Poland.
| |
Collapse
|
27
|
Erbe LS, Hoffjan S, Janßen S, Kneifel M, Krause K, Gerding WM, Döring K, Güttsches AK, Roos A, Buena Atienza E, Gross C, Lücke T, Nguyen HHP, Vorgerd M, Köhler C. Exome Sequencing and Optical Genome Mapping in Molecularly Unsolved Cases of Duchenne Muscular Dystrophy: Identification of a Causative X-Chromosomal Inversion Disrupting the DMD Gene. Int J Mol Sci 2023; 24:14716. [PMID: 37834164 PMCID: PMC10572545 DOI: 10.3390/ijms241914716] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe progressive muscle disease that mainly affects boys due to X-linked recessive inheritance. In most affected individuals, MLPA or sequencing-based techniques detect deletions, duplications, or point mutations in the dystrophin-encoding DMD gene. However, in a small subset of patients clinically diagnosed with DMD, the molecular cause is not identified with these routine methods. Evaluation of the 60 DMD patients in our center revealed three cases without a known genetic cause. DNA samples of these patients were analyzed using whole-exome sequencing (WES) and, if unconclusive, optical genome mapping (OGM). WES led to a diagnosis in two cases: one patient was found to carry a splice mutation in the DMD gene that had not been identified during previous Sanger sequencing. In the second patient, we detected two variants in the fukutin gene (FKTN) that were presumed to be disease-causing. In the third patient, WES was unremarkable, but OGM identified an inversion disrupting the DMD gene (~1.28 Mb) that was subsequently confirmed with long-read sequencing. These results highlight the importance of reanalyzing unsolved cases using WES and demonstrate that OGM is a useful method for identifying large structural variants in cases with unremarkable exome sequencing.
Collapse
Affiliation(s)
- Leoni S. Erbe
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Sabine Hoffjan
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
| | - Sören Janßen
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Moritz Kneifel
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Karsten Krause
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Wanda M. Gerding
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Kristina Döring
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
| | - Anne-Katrin Güttsches
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Andreas Roos
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Elena Buena Atienza
- Institute of Medical Genetics and Applied Genomics, University Tübingen, 72074 Tübingen, Germany; (E.B.A.); (C.G.)
- NGS Competence Center Tübingen, 72076 Tübingen, Germany
| | - Caspar Gross
- Institute of Medical Genetics and Applied Genomics, University Tübingen, 72074 Tübingen, Germany; (E.B.A.); (C.G.)
- NGS Competence Center Tübingen, 72076 Tübingen, Germany
| | - Thomas Lücke
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Hoa Huu Phuc Nguyen
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany; (L.S.E.); (W.M.G.); (K.D.); (H.H.P.N.)
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44801 Bochum, Germany; (M.K.); (K.K.); (A.-K.G.); (A.R.); (M.V.)
| | - Cornelia Köhler
- Center for Rare Diseases Ruhr (CeSER), 44791 Bochum, Germany; (C.K.); (T.L.)
- Department of Neuropediatrics, University Children’s Hospital, Ruhr-University Bochum, 44801 Bochum, Germany;
| |
Collapse
|
28
|
Egorova TV, Polikarpova AV, Vassilieva SG, Dzhenkova MA, Savchenko IM, Velyaev OA, Shmidt AA, Soldatov VO, Pokrovskii MV, Deykin AV, Bardina MV. CRISPR-Cas9 correction in the DMD mouse model is accompanied by upregulation of Dp71f protein. Mol Ther Methods Clin Dev 2023; 30:161-180. [PMID: 37457303 PMCID: PMC10339130 DOI: 10.1016/j.omtm.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe hereditary disease caused by a deficiency in the dystrophin protein. The most frequent types of disease-causing mutations in the DMD gene are frameshift deletions of one or more exons. Precision genome editing systems such as CRISPR-Cas9 have shown potential to restore open reading frames in numerous animal studies. Here, we applied an AAV-CRISPR double-cut strategy to correct a mutation in the DMD mouse model with exon 8-34 deletion, encompassing the N-terminal actin-binding domain. We report successful excision of the 100-kb genomic sequence, which includes exons 6 and 7, and partial improvement in cardiorespiratory function. While corrected mRNA was abundant in muscle tissues, only a low level of truncated dystrophin was produced, possibly because of protein instability. Furthermore, CRISPR-Cas9-mediated genome editing upregulated the Dp71f dystrophin isoform on the sarcolemma. Given the previously reported Dp71-associated muscle pathology, our results question the applicability of genome editing strategies for some DMD patients with N-terminal mutations. The safety and efficacy of CRISPR-Cas9 constructs require rigorous investigation in patient-specific animal models.
Collapse
Affiliation(s)
- Tatiana V. Egorova
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
- Marlin Biotech LLC, Sochi 354340, Russia
| | - Anna V. Polikarpova
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
- Marlin Biotech LLC, Sochi 354340, Russia
| | - Svetlana G. Vassilieva
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Marina A. Dzhenkova
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Irina M. Savchenko
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology Russian Academy of Sciences, Moscow 119334, Russia
| | - Oleg A. Velyaev
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Anna A. Shmidt
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology Russian Academy of Sciences, Moscow 119334, Russia
| | - Vladislav O. Soldatov
- Research Institute of Living Systems Pharmacology, Belgorod National Research University, Belgorod 308007, Russia
| | - Mikhail V. Pokrovskii
- Research Institute of Living Systems Pharmacology, Belgorod National Research University, Belgorod 308007, Russia
| | - Alexey V. Deykin
- Marlin Biotech LLC, Sochi 354340, Russia
- Joint Center for Genetic Technologies, Laboratory of Genetic Technologies and Gene Editing for Biomedicine and Veterinary Medicine, Department of Pharmacology and Clinical Pharmacology, Belgorod National Research University, Belgorod 308015, Russia
| | - Maryana V. Bardina
- Laboratory of Modeling and Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
- Marlin Biotech LLC, Sochi 354340, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
29
|
Wijekoon N, Gonawala L, Ratnayake P, Amaratunga D, Hathout Y, Mohan C, Steinbusch HWM, Dalal A, Hoffman EP, de Silva KRD. Duchenne Muscular Dystrophy from Brain to Muscle: The Role of Brain Dystrophin Isoforms in Motor Functions. J Clin Med 2023; 12:5637. [PMID: 37685704 PMCID: PMC10488491 DOI: 10.3390/jcm12175637] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/26/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Brain function and its effect on motor performance in Duchenne muscular dystrophy (DMD) is an emerging concept. The present study explored how cumulative dystrophin isoform loss, age, and a corticosteroid treatment affect DMD motor outcomes. A total of 133 genetically confirmed DMD patients from Sri Lanka were divided into two groups based on whether their shorter dystrophin isoforms (Dp140, Dp116, and Dp71) were affected: Group 1, containing patients with Dp140, Dp116, and Dp71 affected (n = 98), and Group 2, containing unaffected patients (n = 35). A subset of 52 patients (Group 1, n = 38; Group 2, n = 14) was followed for up to three follow-ups performed in an average of 28-month intervals. The effect of the cumulative loss of shorter dystrophin isoforms on the natural history of DMD was analyzed. A total of 74/133 (56%) patients encountered developmental delays, with 66/74 (89%) being in Group 1 and 8/74 (11%) being in Group 2 (p < 0.001). Motor developmental delays were predominant. The hip and knee muscular strength, according to the Medical Research Council (MRC) scale and the North Star Ambulatory Assessment (NSAA) activities, "standing on one leg R", "standing on one leg L", and "walk", declined rapidly in Group 1 (p < 0.001 In the follow-up analysis, Group 1 patients became wheelchair-bound at a younger age than those of Group 2 (p = 0.004). DMD motor dysfunction is linked to DMD mutations that affect shorter dystrophin isoforms. When stratifying individuals for clinical trials, considering the DMD mutation site and its impact on a shorter dystrophin isoform is crucial.
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; (N.W.); (L.G.)
- 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; (N.W.); (L.G.)
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6200 Maastricht, The Netherlands;
| | | | | | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (Y.H.); (E.P.H.)
| | - Chandra Mohan
- Department of Bioengineering, University of Houston, Houston, TX 77204, 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, Center for DNA Fingerprinting and Diagnostics, Hyderabad 500039, India;
| | - Eric P. Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA; (Y.H.); (E.P.H.)
| | - 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; (N.W.); (L.G.)
- 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
|
30
|
Wijekoon N, Gonawala L, Ratnayake P, Dissanayaka P, Gunarathne I, Amaratunga D, Liyanage R, Senanayaka S, Wijesekara S, Gunasekara HH, Vanarsa K, Castillo J, Hathout Y, Dalal A, Steinbusch HW, Hoffman E, Mohan C, de Silva KRD. Integrated genomic, proteomic and cognitive assessment in Duchenne Muscular Dystrophy suggest astrocyte centric pathology. Heliyon 2023; 9:e18530. [PMID: 37593636 PMCID: PMC10432191 DOI: 10.1016/j.heliyon.2023.e18530] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/15/2023] [Accepted: 07/20/2023] [Indexed: 08/19/2023] Open
Abstract
Introduction Documented Duchenne Muscular Dystrophy (DMD) biomarkers are confined to Caucasians and are poor indicators of cognitive difficulties and neuropsychological alterations. Materials and methods This study correlates serum protein signatures with cognitive performance in DMD patients of South Asian origin. Study included 25 DMD patients aged 6-16 years. Cognitive profiles were assessed by Wechsler Intelligence Scale for Children. Serum proteome profiling of 1317 proteins was performed in eight DMD patients and eight age-matched healthy volunteers. Results Among the several novel observations we report, better cognitive performance in DMD was associated with increased serum levels of MMP9 and FN1 but decreased Siglec-3, C4b, and C3b. Worse cognitive performance was associated with increased serum levels of LDH-H1 and PDGF-BB but reduced GDF-11, MMP12, TPSB2, and G1B. Secondly, better cognitive performance in Processing Speed (PSI) and Perceptual Reasoning (PRI) domains was associated with intact Dp116, Dp140, and Dp71 dystrophin isoforms while better performance in Verbal Comprehension (VCI) and Working Memory (WMI) domains was associated with intact Dp116 and Dp140 isoforms. Finally, functional pathways shared with Alzheimer's Disease (AD) point towards an astrocyte-centric model for DMD. Conclusion Astrocytic dysfunction leading to synaptic dysfunction reported previously in AD may be a common pathogenic mechanism underlying both AD and DMD, linking protein alterations to cognitive impairment. This new insight may pave the path towards novel therapeutic approaches targeting reactive astrocytes.
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, 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, Maastricht, The Netherlands
| | | | - Pulasthi Dissanayaka
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | - Isuru Gunarathne
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | | | - Roshan Liyanage
- Interdisciplinary Center for Innovation in Biotechnology and Neuroscience, Faculty of Medical Sciences, University of Sri Jayewardenepura, Nugegoda, 10250, Sri Lanka
| | | | - Saraji Wijesekara
- Department of Pediatrics, University of Sri Jayewardenepura, 10250, Sri Lanka
- Colombo South Teaching Hospital, 10350, Sri Lanka
| | | | - Kamala Vanarsa
- Department of Bioengineering, University of Houston, Houston, 77204, USA
| | - Jessica Castillo
- Department of Bioengineering, University of Houston, Houston, 77204, USA
| | - Yetrib Hathout
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, USA
| | - Ashwin Dalal
- Diagnostics Division, Center for DNA Fingerprinting and Diagnostics, India
| | - Harry W.M. Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Eric Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, New York, USA
| | - Chandra Mohan
- Department of Bioengineering, University of Houston, Houston, 77204, 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, Maastricht, The Netherlands
- Institute for Combinatorial Advanced Research and Education (KDU-CARE), General Sir John Kotelawala Defence University, Ratmalana, 10390, Sri Lanka
| |
Collapse
|
31
|
Hildyard JCW, Piercy RJ. When Size Really Matters: The Eccentricities of Dystrophin Transcription and the Hazards of Quantifying mRNA from Very Long Genes. Biomedicines 2023; 11:2082. [PMID: 37509720 PMCID: PMC10377302 DOI: 10.3390/biomedicines11072082] [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: 06/27/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
At 2.3 megabases in length, the dystrophin gene is enormous: transcription of a single mRNA requires approximately 16 h. Principally expressed in skeletal muscle, the dystrophin protein product protects the muscle sarcolemma against contraction-induced injury, and dystrophin deficiency results in the fatal muscle-wasting disease, Duchenne muscular dystrophy. This gene is thus of key clinical interest, and therapeutic strategies aimed at eliciting dystrophin restoration require quantitative analysis of its expression. Approaches for quantifying dystrophin at the protein level are well-established, however study at the mRNA level warrants closer scrutiny: measured expression values differ in a sequence-dependent fashion, with significant consequences for data interpretation. In this manuscript, we discuss these nuances of expression and present evidence to support a transcriptional model whereby the long transcription time is coupled to a short mature mRNA half-life, with dystrophin transcripts being predominantly nascent as a consequence. We explore the effects of such a model on cellular transcriptional dynamics and then discuss key implications for the study of dystrophin gene expression, focusing on both conventional (qPCR) and next-gen (RNAseq) approaches.
Collapse
Affiliation(s)
- John C. W. Hildyard
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Science and Services, Royal Veterinary College, London NW1 0TU, UK;
| | | |
Collapse
|
32
|
Elhawary NA, AlJahdali IA, Abumansour IS, Azher ZA, Falemban AH, Madani WM, Alosaimi W, Alghamdi G, Sindi IA. Phenotypic variability to medication management: an update on fragile X syndrome. Hum Genomics 2023; 17:60. [PMID: 37420260 PMCID: PMC10329374 DOI: 10.1186/s40246-023-00507-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
This review discusses the discovery, epidemiology, pathophysiology, genetic etiology, molecular diagnosis, and medication-based management of fragile X syndrome (FXS). It also highlights the syndrome's variable expressivity and common comorbid and overlapping conditions. FXS is an X-linked dominant disorder associated with a wide spectrum of clinical features, including but not limited to intellectual disability, autism spectrum disorder, language deficits, macroorchidism, seizures, and anxiety. Its prevalence in the general population is approximately 1 in 5000-7000 men and 1 in 4000-6000 women worldwide. FXS is associated with the fragile X messenger ribonucleoprotein 1 (FMR1) gene located at locus Xq27.3 and encodes the fragile X messenger ribonucleoprotein (FMRP). Most individuals with FXS have an FMR1 allele with > 200 CGG repeats (full mutation) and hypermethylation of the CpG island proximal to the repeats, which silences the gene's promoter. Some individuals have mosaicism in the size of the CGG repeats or in hypermethylation of the CpG island, both produce some FMRP and give rise to milder cognitive and behavioral deficits than in non-mosaic individuals with FXS. As in several monogenic disorders, modifier genes influence the penetrance of FMR1 mutations and FXS's variable expressivity by regulating the pathophysiological mechanisms related to the syndrome's behavioral features. Although there is no cure for FXS, prenatal molecular diagnostic testing is recommended to facilitate early diagnosis. Pharmacologic agents can reduce some behavioral features of FXS, and researchers are investigating whether gene editing can be used to demethylate the FMR1 promoter region to improve patient outcomes. Moreover, clustered regularly interspaced palindromic repeats (CRISPR)/Cas9 and developed nuclease defective Cas9 (dCas9) strategies have promised options of genome editing in gain-of-function mutations to rewrite new genetic information into a specified DNA site, are also being studied.
Collapse
Affiliation(s)
- Nasser A. Elhawary
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Imad A. AlJahdali
- Department of Community Medicine, College of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Iman S. Abumansour
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Zohor A. Azher
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Alaa H. Falemban
- Department of Pharmacology and Toxicology, College of Medicine, Umm Al-Qura University, Mecca, 24382 Saudi Arabia
| | - Wefaq M. Madani
- Department of Hematology and Immunology, Faculty of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Wafaa Alosaimi
- Department of Hematology, Maternity and Children Hospital, Mecca, Saudi Arabia
| | - Ghydda Alghamdi
- Department of Medical Genetics, College of Medicine, Umm Al-Qura University, Mecca, 21955 Saudi Arabia
| | - Ikhlas A. Sindi
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, 21589 Saudi Arabia
- Preparatory Year Program, Batterjee Medical College, Jeddah, 21442 Saudi Arabia
| |
Collapse
|
33
|
Chrastina J, Haroková M. End of Life in Boys and Young Men With Duchenne Muscular Dystrophy - The Perspective of Dying Men and Their Families: A Systematic Review and Thematic Synthesis of Qualitative Evidence. OMEGA-JOURNAL OF DEATH AND DYING 2023:302228231186358. [PMID: 37408104 DOI: 10.1177/00302228231186358] [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: 07/07/2023]
Abstract
Understanding the perceptions and experiences related to the end of life (EoL) of boys and men with Duchenne muscular dystrophy from their own and/or family perspective is limited based on the available qualitative empirical studies. This systematic review was done with a thematic synthesis of qualitative evidence according to the PRISMA Statement Guidelines and the SPIDER search tool. The review included empirical, qualitative, and relevant full-text studies published in 2000-2023 in the EBSCO Discovery Service, ISI Web of Science, Scopus, PubMed, and ProQuest databases. From o total of eight included qualitative studies, four main key themes were identified: "Being a parent/caregiver" - psychosocial aspects, needs, and experiences; "Communication about EoL with healthcare and other professionals" - positive experiences and personal shortcomings; "Discussions about…" - the issue of EoL, dying and death; and "End of life" - end-of-life care, planning and the need for palliative care.
Collapse
Affiliation(s)
- Jan Chrastina
- Institute of Special Education Studies, Faculty of Education, Palacký University Olomouc, Olomouc, Czech Republic
| | - Martina Haroková
- Institute of Special Education Studies, Faculty of Education, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
34
|
Zhang XF, Luo YY, Jiang L, Hong SQ. Clinical study on cognitive impairment in Duchenne muscular dystrophy. Neuromuscul Disord 2023; 33:596-604. [PMID: 37385106 DOI: 10.1016/j.nmd.2023.06.001] [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: 12/06/2022] [Revised: 05/29/2023] [Accepted: 06/01/2023] [Indexed: 07/01/2023]
Abstract
Our study aimed to explore the intellectual function of patients with Duchenne muscular dystrophy (DMD) in China and examine the correlation of full-scale intelligence quotient (FSIQ) with age, mutation locations, mutation class, and dystrophin isoforms. We assessed 64 boys with DMD using The Wechsler Intelligence Scales for Children-Fourth Edition and compared intellectual function at enrollment and follow-up in the 15 patients who completed the follow-up. Our findings confirm that boys with DMD may exhibit cognitive impairment, with the Working Memory Index being the most impaired. There was no significant correlation between FSIQ and age; however, a positive correlation was noted between age and the Verbal Comprehension Index. FSIQ was not associated with mutation class, the number of affected mutated exons, or mutation locations. However, there was a significant difference in FSIQ between the groups with intact and deficient Dp140. Fifteen participants adhered to glucocorticoid therapy throughout the two-year follow-up period, and eleven of them showed an improvement in FSIQ compared to their initial scores, with improvement ranging from 2 to 20. In conclusion, patients with the cumulative loss of isoforms in the brain are at a higher risk of cognitive deficits and may require early cognitive interventions.
Collapse
Affiliation(s)
- Xiao-Fang Zhang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yuan-Yuan Luo
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Si-Qi Hong
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation base of Child development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
35
|
Donandt T, Todorow V, Hintze S, Graupner A, Schoser B, Walter MC, Meinke P. Nuclear Small Dystrophin Isoforms during Muscle Differentiation. Life (Basel) 2023; 13:1367. [PMID: 37374149 DOI: 10.3390/life13061367] [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: 05/04/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Mutations in the DMD gene can cause Duchenne or Becker muscular dystrophy (DMD/BMD) by affecting the giant isoform of dystrophin, a protein encoded by the DMD gene. The role of small dystrophin isoforms is not well investigated yet, and they may play a role in muscle development and molecular pathology. Here, we investigated the nuclear localization of short carboxy-terminal dystrophin isoforms during the in vitro differentiation of human, porcine, and murine myoblast cultures. We could not only confirm the presence of Dp71 in the nucleoplasm and at the nuclear envelope, but we could also identify the Dp40 isoform in muscle nuclei. The localization of both isoforms over the first six days of differentiation was similar between human and porcine myoblasts, but murine myoblasts behaved differently. This highlights the importance of the porcine model in investigating DMD. We could also detect a wave-like pattern of nuclear presence of both Dp71 and Dp40, indicating a direct or indirect involvement in gene expression control during muscle differentiation.
Collapse
Affiliation(s)
- Tina Donandt
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Vanessa Todorow
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Stefan Hintze
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Alexandra Graupner
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Benedikt Schoser
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| | - Peter Meinke
- Friedrich-Baur-Institute at the Department of Neurology, LMU University Hospital, Ludwig Maximilians University, 81377 Munich, Germany
| |
Collapse
|
36
|
Tang JM, McClennan A, Liu L, Hadway J, Ronald JA, Hicks JW, Hoffman L, Anazodo UC. A Protocol for Simultaneous In Vivo Imaging of Cardiac and Neuroinflammation in Dystrophin-Deficient MDX Mice Using [ 18F]FEPPA PET. Int J Mol Sci 2023; 24:ijms24087522. [PMID: 37108685 PMCID: PMC10144317 DOI: 10.3390/ijms24087522] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder caused by dystrophin loss-notably within muscles and the central neurons system. DMD presents as cognitive weakness, progressive skeletal and cardiac muscle degeneration until pre-mature death from cardiac or respiratory failure. Innovative therapies have improved life expectancy; however, this is accompanied by increased late-onset heart failure and emergent cognitive degeneration. Thus, better assessment of dystrophic heart and brain pathophysiology is needed. Chronic inflammation is strongly associated with skeletal and cardiac muscle degeneration; however, neuroinflammation's role is largely unknown in DMD despite being prevalent in other neurodegenerative diseases. Here, we present an inflammatory marker translocator protein (TSPO) positron emission tomography (PET) protocol for in vivo concomitant assessment of immune cell response in hearts and brains of a dystrophin-deficient mouse model [mdx:utrn(+/-)]. Preliminary analysis of whole-body PET imaging using the TSPO radiotracer, [18F]FEPPA in four mdx:utrn(+/-) and six wildtype mice are presented with ex vivo TSPO-immunofluorescence tissue staining. The mdx:utrn(+/-) mice showed significant elevations in heart and brain [18F]FEPPA activity, which correlated with increased ex vivo fluorescence intensity, highlighting the potential of TSPO-PET to simultaneously assess presence of cardiac and neuroinflammation in dystrophic heart and brain, as well as in several organs within a DMD model.
Collapse
Affiliation(s)
- Joanne M Tang
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Andrew McClennan
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Linshan Liu
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Jennifer Hadway
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - John A Ronald
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Robarts Research Institute, Western University, London, ON N6A 3K7, Canada
| | - Justin W Hicks
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
| | - Lisa Hoffman
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
- Department of Anatomy and Cell Biology, Western University, London, ON N6A 3K7, Canada
| | - Udunna C Anazodo
- Department of Medical Biophysics, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 4V2, Canada
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
37
|
Lindsay A, Russell AP. The unconditioned fear response in dystrophin-deficient mice is associated with adrenal and vascular function. Sci Rep 2023; 13:5513. [PMID: 37015991 PMCID: PMC10073118 DOI: 10.1038/s41598-023-32163-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/23/2023] [Indexed: 04/06/2023] Open
Abstract
Loss of function mutations in the gene encoding dystrophin elicits a hypersensitive fear response in mice and humans. In the dystrophin-deficient mdx mouse, this behaviour is partially protected by oestrogen, but the mechanistic basis for this protection is unknown. Here, we show that female mdx mice remain normotensive during restraint stress compared to a hypotensive and hypertensive response in male mdx and male/female wildtype mice, respectively. Partial dystrophin expression in female mdx mice (heterozygous) also elicited a hypertensive response. Ovariectomized (OVX) female mdx mice were used to explain the normotensive response to stress. OVX lowered skeletal muscle mass and lowered the adrenal mass and zona glomerulosa area (aldosterone synthesis) in female mdx mice. During a restraint stress, OVX dampened aldosterone synthesis and lowered the corticosterone:11-dehydrocorticosterone. All OVX-induced changes were restored with replacement of oestradiol, except that oestradiol lowered the zona fasciculata area of the adrenal gland, dampened corticosterone synthesis but increased cortisol synthesis. These data suggest that oestrogen partially attenuates the unconditioned fear response in mdx mice via adrenal and vascular function. It also suggests that partial dystrophin restoration in a dystrophin-deficient vertebrate is an effective approach to develop an appropriate hypertensive response to stress.
Collapse
Affiliation(s)
- Angus Lindsay
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia.
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| |
Collapse
|
38
|
Dystrophin Short Product, Dp71, Interacts with AQP4 and Kir4.1 Channels in the Mouse Cerebellar Glial Cells in Contrast to Dp427 at Inhibitory Postsynapses in the Purkinje Neurons. Mol Neurobiol 2023; 60:3664-3677. [PMID: 36918517 DOI: 10.1007/s12035-023-03296-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 03/01/2023] [Indexed: 03/16/2023]
Abstract
Dystrophin is the causative gene for Duchenne and Becker muscular dystrophy (DMD/BMD), and it produces full-length and short dystrophin, Dp427 and Dp71, respectively, in the brain. The existence of the different dystrophin molecular complexes has been known for a quarter century, so it is necessary to derive precise expression profiles of the molecular complexes in the brain to elucidate the mechanism of cognitive symptoms in DMD/BMD patients. In order to investigate the Dp71 expression profile in cerebellum, we employed Dp71-specific tag-insertion mice, which allowed for the specific detection of endogenous Dp71 in the immunohistochemical analysis and found its expressions in the glial cells, Bergmann glial (BG) cells, and astrocytes, whereas Dp427 was exclusively expressed in the inhibitory postsynapses within cerebellar Purkinje cells (PCs). Interestingly, we found different cell-type dependent dystrophin molecular complexes; i.e., glia-associated Dp71 was co-expressed with dystroglycan (DG) and dystrobrevinα, whereas synapse-associated Dp427 was co-expressed with DG and dystrobrevinβ. Furthermore, we investigated the molecular relationship of Dp71 to the AQP4 water channel and the Kir4.1 potassium channel, and found biochemical associations of Dp71 with AQP4 and Kir4.1 in both the cerebellum and cerebrum. Immunohistochemical and cytochemical investigations revealed partial co-localizations of Dp71 with AQP4 and Kir4.1 in the glial cells, indicating Dp71 interactions with the channels in the BG cells and astrocytes. Taken together, different cell-types, glial cells and Purkinje neurons, in the cerebellum express different dystrophin molecular complexes, which may contribute to pathological and physiological processes through the regulation of the water/ion channel and inhibitory postsynapses.
Collapse
|
39
|
Downregulation of Dystrophin Expression Occurs across Diverse Tumors, Correlates with the Age of Onset, Staging and Reduced Survival of Patients. Cancers (Basel) 2023; 15:cancers15051378. [PMID: 36900171 PMCID: PMC10000051 DOI: 10.3390/cancers15051378] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
Altered dystrophin expression was found in some tumors and recent studies identified a developmental onset of Duchenne muscular dystrophy (DMD). Given that embryogenesis and carcinogenesis share many mechanisms, we analyzed a broad spectrum of tumors to establish whether dystrophin alteration evokes related outcomes. Transcriptomic, proteomic, and mutation datasets from fifty tumor tissues and matching controls (10,894 samples) and 140 corresponding tumor cell lines were analyzed. Interestingly, dystrophin transcripts and protein expression were found widespread across healthy tissues and at housekeeping gene levels. In 80% of tumors, DMD expression was reduced due to transcriptional downregulation and not somatic mutations. The full-length transcript encoding Dp427 was decreased in 68% of tumors, while Dp71 variants showed variability of expression. Notably, low expression of dystrophins was associated with a more advanced stage, older age of onset, and reduced survival across different tumors. Hierarchical clustering analysis of DMD transcripts distinguished malignant from control tissues. Transcriptomes of primary tumors and tumor cell lines with low DMD expression showed enrichment of specific pathways in the differentially expressed genes. Pathways consistently identified: ECM-receptor interaction, calcium signaling, and PI3K-Akt are also altered in DMD muscle. Therefore, the importance of this largest known gene extends beyond its roles identified in DMD, and certainly into oncology.
Collapse
|
40
|
Shirokova O, Zaborskaya O, Pchelin P, Kozliaeva E, Pershin V, Mukhina I. Genetic and Epigenetic Sexual Dimorphism of Brain Cells during Aging. Brain Sci 2023; 13:brainsci13020195. [PMID: 36831738 PMCID: PMC9954625 DOI: 10.3390/brainsci13020195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
In recent years, much of the attention paid to theoretical and applied biomedicine, as well as neurobiology, has been drawn to various aspects of sexual dimorphism due to the differences that male and female brain cells demonstrate during aging: (a) a dimorphic pattern of response to therapy for neurodegenerative disorders, (b) different age of onset and different degrees of the prevalence of such disorders, and (c) differences in their symptomatic manifestations in men and women. The purpose of this review is to outline the genetic and epigenetic differences in brain cells during aging in males and females. As a result, we hereby show that the presence of brain aging patterns in males and females is due to a complex of factors associated with the effects of sex chromosomes, which subsequently entails a change in signal cascades in somatic cells.
Collapse
Affiliation(s)
- Olesya Shirokova
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Correspondence:
| | - Olga Zaborskaya
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
| | - Pavel Pchelin
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| | - Elizaveta Kozliaeva
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
| | - Vladimir Pershin
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| | - Irina Mukhina
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod 603950, Russia
- Institute of Biology and Biomedicine, Lobachevsky State University, 23 Gagarin Avenue, Nizhny Novgorod 603002, Russia
| |
Collapse
|
41
|
Maresh K, Papageorgiou A, Ridout D, Harrison NA, Mandy W, Skuse D, Muntoni F. Startle responses in Duchenne muscular dystrophy: a novel biomarker of brain dystrophin deficiency. Brain 2023; 146:252-265. [PMID: 35136951 PMCID: PMC9825594 DOI: 10.1093/brain/awac048] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/20/2021] [Accepted: 01/16/2022] [Indexed: 01/12/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by loss of dystrophin in muscle, however patients also have variable degree of intellectual disability and neurobehavioural co-morbidities. In contrast to muscle, in which a single full-length dystrophin isoform (Dp427) is produced, multiple isoforms are produced in the brain, and their deficiency accounts for the variability of CNS manifestations, with increased risk of comorbidities in patients carrying mutations affecting the 3' end of the gene, which disrupt expression of shorter Dp140 and Dp71 isoforms. A mouse model (mdx mouse) lacks Dp427 in muscle and CNS and exhibits exaggerated startle responses to threat, linked to the deficiency of dystrophin in limbic structures such as the amygdala, which normalize with postnatal brain dystrophin-restoration therapies. A pathological startle response is not a recognized feature of DMD, and its characterization has implications for improved clinical management and translational research. To investigate startle responses in DMD, we used a novel fear-conditioning task in an observational study of 56 males aged 7-12 years (31 affected boys, mean age 9.7 ± 1.8 years; 25 controls, mean age 9.6 ± 1.4 years). Trials of two neutral visual stimuli were presented to participants: one 'safe' cue presented alone; one 'threat' cue paired with an aversive noise to enable conditioning of physiological startle responses (skin conductance response and heart rate). Retention of conditioned physiological responses was subsequently tested by presenting both cues without the aversive noise in an 'Extinction' phase. Primary outcomes were the initial unconditioned skin conductance and change in heart rate responses to the aversive 'threat' and acquisition and retention of conditioned responses after conditioning. Secondary and exploratory outcomes were neuropsychological measures and genotype associations. The mean unconditioned skin conductance response was greater in the DMD group than controls [mean difference 3.0 µS (1.0, 5.1); P = 0.004], associated with a significant threat-induced bradycardia only in the patient group [mean difference -8.7 bpm (-16.9, -0.51); P = 0.04]. Participants with DMD found the task more aversive than controls, with increased early termination rates during the Extinction phase (26% of DMD group versus 0% of controls; P = 0.007). This study provides the first evidence that boys with DMD show similar increased unconditioned startle responses to threat to the mdx mouse, which in the mouse respond to brain dystrophin restoration. Our study provides new insights into the neurobiology underlying the complex neuropsychiatric co-morbidities in DMD and defines an objective measure of this CNS phenotype, which will be valuable for future CNS-targeted dystrophin-restoration studies.
Collapse
Affiliation(s)
- Kate Maresh
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Queen Square Centre for Neuromuscular Diseases, University College London, London WC1N 3BG, UK
| | - Andriani Papageorgiou
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Deborah Ridout
- Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Neil A Harrison
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - William Mandy
- Department of Clinical, Educational and Health Psychology, University College London, London WC1E 6BT, UK
| | - David Skuse
- Department of Behavioural and Brain Sciences, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Queen Square Centre for Neuromuscular Diseases, University College London, London WC1N 3BG, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| |
Collapse
|
42
|
Chieffo DPR, Moriconi F, Pane M, Lucibello S, Ferraroli E, Norcia G, Ricci M, Capasso A, Cicala G, Buchignani B, Coratti G, Cutrona C, Pelizzari M, Brogna C, Hendriksen JGM, Muntoni F, Mercuri E. A Longitudinal Follow-Up Study of Intellectual Function in Duchenne Muscular Dystrophy over Age: Is It Really Stable? J Clin Med 2023; 12:jcm12020403. [PMID: 36675332 PMCID: PMC9865074 DOI: 10.3390/jcm12020403] [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: 11/29/2022] [Revised: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
The aim of the study was to retrospectively evaluate the consistency of longitudinal findings on intellectual functioning in DMD boys and their relationship to behavioral and neuropsychiatric difficulties. The cohort included 70 patients of age 3 to 17 years with at least two assessments using the Wechsler scales. CBCL and clinical observation of behavior were also performed. Changes in total intelligence quotient were interpreted as stable or not stable using the reliable-change method. On the first assessment 43/70 had normal quotients, 18 borderline, 5 mild, and 4 moderate intellectual disability, while 27/70 had no behavioral disorders, 17 had abnormal CBCL, and 26 patients had clear signs of attention deficits despite normal CBCL. The remaining seven were untestable. The mean total intelligence quotient change in the cohort was -2.99 points (SD: 12.29). Stable results on TIQ were found in 63% of the paired assessments. A third of the consecutive cognitive assessments showed a difference of more than 11 points with changes up to 42 points. Boys with no behavioral/attention disorder had smaller changes than those with attention (p = 0.007) and behavioral disorders (p = 0.002). Changes in IQ may occur in Duchenne and are likely to be associated with behavioral or attention deficits.
Collapse
Affiliation(s)
- Daniela P. R. Chieffo
- Psychology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Federica Moriconi
- Psychology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Marika Pane
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Simona Lucibello
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Elisabetta Ferraroli
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giulia Norcia
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Martina Ricci
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Anna Capasso
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Gianpaolo Cicala
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Bianca Buchignani
- Department of Clinical and Experimental Medicine, Università di Pisa, 56126 Pisa, Italy
| | - Giorgia Coratti
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Costanza Cutrona
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Monia Pelizzari
- Psychology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Claudia Brogna
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Jos G. M. Hendriksen
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
- Great Ormond Street Hospital Trust, London WC1N 1EH, UK
| | - Eugenio Mercuri
- Department of Life Science and Public Health, Pediatric Neurology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Correspondence: ; Tel.: +39-06-3015-5340
| |
Collapse
|
43
|
Pokrovsky MV, Korokin MV, Krayushkina AM, Zhunusov NS, Lapin KN, Soldatova MO, Kuzmin EA, Gudyrev OS, Kochkarova IS, Deikin AV. CONVENTIONAL APPROACHES TO THE THERAPY OF HEREDITARY MYOPATHIES. PHARMACY & PHARMACOLOGY 2022. [DOI: 10.19163/2307-9266-2022-10-5-416-431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the work was to analyze the available therapeutic options for the conventional therapy of hereditary myopathies.Materials and methods. When searching for the material for writing a review article, such abstract databases as PubMed and Google Scholar were used. The search was carried out on the publications during the period from 1980 to September 2022. The following words and their combinations were selected as parameters for the literature selection: “myopathy”, “Duchenne”, “myodystrophy”, “metabolic”, “mitochondrial”, “congenital”, “symptoms”, “replacement”, “recombinant”, “corticosteroids”, “vitamins”, “tirasemtiv”, “therapy”, “treatment”, “evidence”, “clinical trials”, “patients”, “dichloracetate”.Results. Congenital myopathies are a heterogeneous group of pathologies that are caused by atrophy and degeneration of muscle fibers due to mutations in genes. Based on a number of clinical and pathogenetic features, hereditary myopathies are divided into: 1) congenital myopathies; 2) muscular dystrophy; 3) mitochondrial and 4) metabolic myopathies. At the same time, treatment approaches vary significantly depending on the type of myopathy and can be based on 1) substitution of the mutant protein; 2) an increase in its expression; 3) stimulation of the internal compensatory pathways expression; 4) restoration of the compounds balance associated with the mutant protein function (for enzymes); 5) impact on the mitochondrial function (with metabolic and mitochondrial myopathies); 6) reduction of inflammation and fibrosis (with muscular dystrophies); as well as 7) an increase in muscle mass and strength. The current review presents current data on each of the listed approaches, as well as specific pharmacological agents with a description of their action mechanisms.Conclusion. Currently, the following pharmacological groups are used or undergoing clinical trials for the treatment of various myopathies types: inotropic, anti-inflammatory and antifibrotic drugs, antimyostatin therapy and the drugs that promote translation through stop codons (applicable for nonsense mutations). In addition, metabolic drugs, metabolic enzyme cofactors, mitochondrial biogenesis stimulators, and antioxidants can be used to treat myopathies. Finally, the recombinant drugs alglucosidase and avalglucosidase have been clinically approved for the replacement therapy of metabolic myopathies (Pompe’s disease).
Collapse
Affiliation(s)
| | | | | | | | - K. N. Lapin
- V.A. Negovsky Research Institute of General Reanimatology, Federal Scientific and Clinical Center for Resuscitation and Rehabilitology
| | | | - E. A. Kuzmin
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | |
Collapse
|
44
|
Pascual-Morena C, Cavero-Redondo I, Álvarez-Bueno C, Jiménez-López E, Saz-Lara A, Martínez-García I, Martínez-Vizcaíno V. Global prevalence of intellectual developmental disorder in dystrophinopathies: A systematic review and meta-analysis. Dev Med Child Neurol 2022; 65:734-744. [PMID: 36440509 DOI: 10.1111/dmcn.15481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/16/2022] [Accepted: 11/08/2022] [Indexed: 11/29/2022]
Abstract
AIM To estimate the global prevalence of intellectual developmental disorder (IDD) and the IDD prevalence-genotype association in Becker muscular dystrophy (BMD) or Duchenne muscular dystrophy (DMD) according to the affected isoforms of the DMD gene: Dp427, Dp140, Dp71. METHOD Systematic searches in MEDLINE, Scopus, Web of Science, and the Cochrane Library were conducted from inception of each database to March 2022. Observational studies that determined the prevalence of IDD in the population with BMD or DMD were included. Meta-analyses of IDD prevalence and prevalence ratios of the IDD-genotype association were conducted. RESULTS Forty-nine studies were included. The prevalence of IDD in BMD was 8.0% (95% confidence interval 5.0-11.0), and in DMD it was 22.0% (18.0-27.0). Meta-analyses of IDD-genotype association showed a deleterious association between IDD and the number of isoforms affected in DMD, with a prevalence ratio = 0.43 (0.28-0.64) and 0.17 (0.09-0.34) for Dp140+ /Dp71+ versus Dp140- /Dp71+ and Dp140+ /Dp71+ versus Dp140- /Dp71- comparisons respectively. However, in BMD, there was no association for Dp140+ /Dp71+ versus Dp140- /Dp71+ . INTERPRETATION There is a high prevalence of IDD in BMD and DMD. Moreover, the number of isoforms affected is strongly and negatively associated with the prevalence of IDD in DMD.
Collapse
Affiliation(s)
| | - Iván Cavero-Redondo
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| | - Celia Álvarez-Bueno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain.,Universidad Politécnica y Artística del Paraguay, Asunción, Paraguay
| | - Estela Jiménez-López
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Alicia Saz-Lara
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain
| | | | - Vicente Martínez-Vizcaíno
- Health and Social Research Center, Universidad de Castilla-La Mancha, Cuenca, Spain.,Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
45
|
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: 8] [Impact Index Per Article: 2.7] [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
|
46
|
Counterman KJ, Fatovic K, Good DC, Martin AS, Dasgupta S, Anziska Y. Associations Between Self-Reported Behavioral and Learning Concerns and DMD Isoforms in Duchenne Muscular Dystrophy. J Neuromuscul Dis 2022; 9:757-764. [DOI: 10.3233/jnd-220821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Duchenne muscular dystrophy (DMD) is an X-linked recessive neuromuscular disorder resulting from loss of dystrophin. In addition to its role in muscle, isoforms of dystrophin are expressed in different cell types of the brain, and DMD has been linked to language delays, behavioral abnormalities and learning disabilities. Objective: To determine whether disruption of specific DMD isoforms, age, corticosteroid use, ambulation status, or country are associated with behavioral and/or learning concerns in DMD. Methods: De-identified data were collected from the Duchenne Registry from 2007– 2019. Females, patients with BMD, and those without genetic testing reports were excluded from the cohort. For the genetic analysis, patients were divided into four subgroups based on the location of their mutation and the predicted isoforms affected. Bivariate analysis was conducted using chi-square for categorical variables. Two multivariate logistic regressions were used to assess independent associations with behavioral and learning concerns, respectively, and to estimate the effect size of each variable. Results: DMD mutations disrupting expression of Dp140 and Dp71 were associated with a higher likelihood of reported behavioral and learning concerns. Corticosteroid use, categorical age, and country were other factors associated with behavior and learning concerns. Conclusion: This data adds to our current understanding of DMD isoforms, their mutational consequence and impact on behavior and learning.
Collapse
Affiliation(s)
| | - Kathy Fatovic
- University of New England College of Osteopathic Medicine, Biddeford, ME
| | - Daniel C. Good
- University of New England College of Osteopathic Medicine, Biddeford, ME
| | | | | | - Yaacov Anziska
- State University of New York Downstate Medical Center, Brooklyn, New York, NY
| |
Collapse
|
47
|
Rugerio-Martínez CI, Ramos D, Segura-Olvera A, Murillo-Melo NM, Tapia-Guerrero YS, Argüello-García R, Leyva-García N, Hernández-Hernández O, Cisneros B, Suárez-Sánchez R. Dp71 Point Mutations Induce Protein Aggregation, Loss of Nuclear Lamina Integrity and Impaired Braf35 and Ibraf Function in Neuronal Cells. Int J Mol Sci 2022; 23:ijms231911876. [PMID: 36233175 PMCID: PMC9570083 DOI: 10.3390/ijms231911876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/04/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Dystrophin Dp71 is the most abundant product of the Duchenne muscular dystrophy gene in the nervous system, and mutations impairing its function have been associated with the neurodevelopmental symptoms present in a third of DMD patients. Dp71 is required for the clustering of neurotransmitter receptors and the neuronal differentiation of cultured cells; nonetheless, its precise role in neuronal cells remains to be poorly understood. In this study, we analyzed the effect of two pathogenic DMD gene point mutations on the Dp71 function in neurons. We engineered C272Y and E299del mutations to express GFP-tagged Dp71 protein variants in N1E-115 and SH-SY5Y neuronal cells. Unexpectedly, the ectopic expression of Dp71 mutants resulted in protein aggregation, which may be mechanistically caused by the effect of the mutations on Dp71 structure, as predicted by protein modeling and molecular dynamics simulations. Interestingly, Dp71 mutant variants acquired a dominant negative function that, in turn, dramatically impaired the distribution of different Dp71 protein partners, including β-dystroglycan, nuclear lamins A/C and B1, the high-mobility group (HMG)-containing protein (BRAF35) and the BRAF35-family-member inhibitor of BRAF35 (iBRAF). Further analysis of Dp71 mutants provided evidence showing a role for Dp71 in modulating both heterochromatin marker H3K9me2 organization and the neuronal genes’ expression, via its interaction with iBRAF and BRAF5.
Collapse
Affiliation(s)
- Claudia Ivette Rugerio-Martínez
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Daniel Ramos
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Abel Segura-Olvera
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Nadia Mireya Murillo-Melo
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Yessica Sarai Tapia-Guerrero
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Raúl Argüello-García
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Norberto Leyva-García
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Oscar Hernández-Hernández
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
| | - Bulmaro Cisneros
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de Mexico 07360, Mexico
| | - Rocío Suárez-Sánchez
- Laboratorio de Medicina Genómica, Departamento de Genética, Instituto Nacional de Rehabilitación-Luis Guillermo Ibarra Ibarra, Ciudad de Mexico 14389, Mexico
- Correspondence: or ; Tel.: +52-55-5999-1000 (ext. 14710)
| |
Collapse
|
48
|
Jackson T, Seifi M, Górecki DC, Swinny JD. Specific Dystrophins Selectively Associate with Inhibitory and Excitatory Synapses of the Mouse Cerebellum and their Loss Alters Expression of P2X7 Purinoceptors and Pro-Inflammatory Mediators. Cell Mol Neurobiol 2022; 42:2357-2377. [PMID: 34101068 PMCID: PMC9418305 DOI: 10.1007/s10571-021-01110-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) patients, having mutations of the DMD gene, present with a range of neuropsychiatric disorders, in addition to the quintessential muscle pathology. The neurobiological basis remains poorly understood because the contributions of different DMD gene products (dystrophins) to the different neural networks underlying such symptoms are yet to be fully characterised. While full-length dystrophin clusters in inhibitory synapses, with inhibitory neurotransmitter receptors, the precise subcellular expression of truncated DMD gene products with excitatory synapses remains unresolved. Furthermore, inflammation, involving P2X purinoceptor 7 (P2RX7) accompanies DMD muscle pathology, yet any association with brain dystrophins is yet to be established. The aim of this study was to investigate the comparative expression of different dystrophins, alongside ionotropic glutamate receptors and P2RX7s, within the cerebellar circuitry known to express different dystrophin isoforms. Immunoreactivity for truncated DMD gene products was targeted to Purkinje cell (PC) distal dendrites adjacent to, or overlapping with, signal for GluA1, GluA4, GluN2A, and GluD2 receptor subunits. P2X7R immunoreactivity was located in Bergmann glia profiles adjacent to PC-dystrophin immunoreactivity. Ablation of all DMD gene products coincided with decreased mRNA expression for Gria2, Gria3, and Grin2a and increased GluD2 immunoreactivity. Finally, dystrophin-null mice showed decreased brain mRNA expression of P2rx7 and several inflammatory mediators. The data suggest that PCs target different dystrophin isoforms to molecularly and functionally distinct populations of synapses. In contrast to muscle, dystrophinopathy in brain leads to the dampening of the local immune system.
Collapse
Affiliation(s)
- Torquil Jackson
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, Portsmouth, PO12DT, UK
| | - Mohsen Seifi
- Leicester School of Pharmacy, De Montfort University, Leicester, LE1 9BH, UK
| | - Dariusz C Górecki
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, Portsmouth, PO12DT, UK
- Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-001, Warsaw, Poland
| | - Jerome D Swinny
- School of Pharmacy & Biomedical Sciences, University of Portsmouth, St Michael's Building, White Swan Road, Portsmouth, PO12DT, UK.
| |
Collapse
|
49
|
The role of the dystrophin glycoprotein complex in muscle cell mechanotransduction. Commun Biol 2022; 5:1022. [PMID: 36168044 PMCID: PMC9515174 DOI: 10.1038/s42003-022-03980-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/12/2022] [Indexed: 11/09/2022] Open
Abstract
Dystrophin is the central protein of the dystrophin-glycoprotein complex (DGC) in skeletal and heart muscle cells. Dystrophin connects the actin cytoskeleton to the extracellular matrix (ECM). Severing the link between the ECM and the intracellular cytoskeleton has a devastating impact on the homeostasis of skeletal muscle cells, leading to a range of muscular dystrophies. In addition, the loss of a functional DGC leads to progressive dilated cardiomyopathy and premature death. Dystrophin functions as a molecular spring and the DGC plays a critical role in maintaining the integrity of the sarcolemma. Additionally, evidence is accumulating, linking the DGC to mechanosignalling, albeit this role is still less understood. This review article aims at providing an up-to-date perspective on the DGC and its role in mechanotransduction. We first discuss the intricate relationship between muscle cell mechanics and function, before examining the recent research for a role of the dystrophin glycoprotein complex in mechanotransduction and maintaining the biomechanical integrity of muscle cells. Finally, we review the current literature to map out how DGC signalling intersects with mechanical signalling pathways to highlight potential future points of intervention, especially with a focus on cardiomyopathies. A review of the function of the Dystrophic Glycoprotein Complex (DGC) in mechanosignaling provides an overview of the various components of DGC and potential mechanopathogenic mechanisms, particularly as they relate to muscular dystrophy.
Collapse
|
50
|
García-Cruz C, Aragón J, Lourdel S, Annan A, Roger JE, Montanez C, Vaillend C. Tissue- and cell-specific whole-transcriptome meta-analysis from brain and retina reveals differential expression of dystrophin complexes and new dystrophin spliced isoforms. Hum Mol Genet 2022; 32:659-676. [PMID: 36130212 PMCID: PMC9896479 DOI: 10.1093/hmg/ddac236] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 02/07/2023] Open
Abstract
The large DMD gene encodes a group of dystrophin proteins in brain and retina, produced from multiple promoters and alternative splicing events. Dystrophins are core components of different scaffolding complexes in distinct cell types. Their absence may thus alter several cellular pathways, which might explain the heterogeneous genotype-phenotype relationships underlying central comorbidities in Duchenne muscular dystrophy (DMD). However, the cell-specific expression of dystrophins and associated proteins (DAPs) is still largely unknown. The present study provides a first RNA-Seq-based reference showing tissue- and cell-specific differential expression of dystrophins, splice variants and DAPs in mouse brain and retina. We report that a cell type may express several dystrophin complexes, perhaps due to expression in separate cell subdomains and/or subpopulations, some of which with differential expression at different maturation stages. We also identified new splicing events in addition to the common exon-skipping events. These include a new exon within intron 51 (E51b) in frame with the flanking exons in retina, as well as inclusions of intronic sequences with stop codons leading to the presence of transcripts with elongated exons 40 and/or 41 (E40e, E41e) in both retina and brain. PCR validations revealed that the new exons may affect several dystrophins. Moreover, immunoblot experiments using a combination of specific antibodies and dystrophin-deficient mice unveiled that the transcripts with stop codons are translated into truncated proteins lacking their C-terminus, which we called N-Dp427 and N-Dp260. This study thus uncovers a range of new findings underlying the complex neurobiology of DMD.
Collapse
Affiliation(s)
| | | | - Sophie Lourdel
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, 91400 Saclay, France
| | - Ahrmad Annan
- Institut des Neurosciences Paris Saclay, Université Paris-Saclay, CNRS, 91400 Saclay, France
| | - Jérôme E Roger
- To whom correspondence should be addressed. E-mail: (C.V.); (C.M.); (J.E.R.)
| | - Cecilia Montanez
- To whom correspondence should be addressed. E-mail: (C.V.); (C.M.); (J.E.R.)
| | - Cyrille Vaillend
- To whom correspondence should be addressed. E-mail: (C.V.); (C.M.); (J.E.R.)
| |
Collapse
|